1
|
Jiang S, Wang P, Sun X, Zhang M, Zhang S, Cao Y, Wang Y, Liu L, Gao X. Mechanistic study of leukopenia treatment by Qijiao shengbai Capsule via the Bcl2/Bax/CASAPSE3 pathway. Front Pharmacol 2024; 15:1451553. [PMID: 39295929 PMCID: PMC11408280 DOI: 10.3389/fphar.2024.1451553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/05/2024] [Indexed: 09/21/2024] Open
Abstract
Background Leukopenia can be caused by chemotherapy, which suppresses bone marrow function and can impact the effectiveness of cancer treatment. Qijiao Shengbai Capsule (QJSB) is commonly used to treat leukopenia, but the specific bioactive components and mechanisms of action are not well understood. Objectives and results This study aimed to analyze the active ingredients of QJSB and its potential targets for treating leukopenia using network pharmacology and molecular docking. Through a combination of serum pharmacochemistry, multi-omics, network pharmacology, and validation experiments in a murine leukopenia model, the researchers sought to understand how QJSB improves leukopenia. The study identified 16 key components of QJSB that act in vivo to increase the number of white blood cells in leukopenic mice. Multi-omics analysis and network pharmacology revealed that the PI3K-Akt and MAPK signaling pathways are important in the treatment of leukopenia with QJSB. Five specific targets (JUN, FOS, BCl-2, CASPAS-3) were identified as key targets. Conclusion Validation experiments confirmed that QJSB regulates genes related to cell growth and inhibits apoptosis, suggesting that apoptosis may play a crucial role in leukopenia development and that QJSB may improve immune function by regulating apoptotic proteins and increasing CD4+ T cell count in leukopenic mice.
Collapse
Affiliation(s)
- Siyue Jiang
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
| | - Pengjiao Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
| | - Xiaodong Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
| | - Min Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
| | - Shuo Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Experimental Animal Center of Guizhou Medical University, Guiyang, China
| | - Yu Cao
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
| | - Yuben Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
| | - Li Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
| | - Xiuli Gao
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang, China
- Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang, China
| |
Collapse
|
2
|
Li J, Huang X, An Y, Chen X, Chen Y, Xu M, Shan H, Zhang M. The role of snapin in regulation of brain homeostasis. Neural Regen Res 2024; 19:1696-1701. [PMID: 38103234 PMCID: PMC10960280 DOI: 10.4103/1673-5374.389364] [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: 05/29/2023] [Revised: 09/15/2023] [Accepted: 10/08/2023] [Indexed: 12/18/2023] Open
Abstract
Brain homeostasis refers to the normal working state of the brain in a certain period, which is important for overall health and normal life activities. Currently, there is a lack of effective treatment methods for the adverse consequences caused by brain homeostasis imbalance. Snapin is a protein that assists in the formation of neuronal synapses and plays a crucial role in the normal growth and development of synapses. Recently, many researchers have reported the association between snapin and neurologic and psychiatric disorders, demonstrating that snapin can improve brain homeostasis. Clinical manifestations of brain disease often involve imbalances in brain homeostasis and may lead to neurological and behavioral sequelae. This article aims to explore the role of snapin in restoring brain homeostasis after injury or diseases, highlighting its significance in maintaining brain homeostasis and treating brain diseases. Additionally, it comprehensively discusses the implications of snapin in other extracerebral diseases such as diabetes and viral infections, with the objective of determining the clinical potential of snapin in maintaining brain homeostasis.
Collapse
Affiliation(s)
- Jiawen Li
- Shanghai Key Lab of Forensic Medicine, Key Lab of Forensic Science, Ministry of Justice, China (Academy of Forensic Science), Shanghai, China
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Xinqi Huang
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Yumei An
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Xueshi Chen
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Yiyang Chen
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Mingyuan Xu
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Haiyan Shan
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Mingyang Zhang
- Shanghai Key Lab of Forensic Medicine, Key Lab of Forensic Science, Ministry of Justice, China (Academy of Forensic Science), Shanghai, China
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
3
|
Cai J, Hu Q, He Z, Chen X, Wang J, Yin X, Ma X, Zeng J. Scutellaria baicalensis Georgi and Their Natural Flavonoid Compounds in the Treatment of Ovarian Cancer: A Review. Molecules 2023; 28:5082. [PMID: 37446743 DOI: 10.3390/molecules28135082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Ovarian cancer (OC) is one of the most common types of cancer in women with a high mortality rate, and the treatment of OC is prone to high recurrence rates and side effects. Scutellaria baicalensis (SB) is a herbal medicine with good anti-cancer activity, and several studies have shown that SB and its flavonoids have some anti-OC properties. This paper elucidated the common pathogenesis of OC, including cell proliferation and cell cycle regulation, cell invasion and metastasis, apoptosis and autophagy, drug resistance and angiogenesis. The mechanisms of SB and its flavonoids, wogonin, baicalein, baicalin, Oroxylin A, and scutellarein, in the treatment of OC, are revealed, such as wogonin inhibits proliferation, induces apoptosis, inhibits invasion and metastasis, and increases the cytotoxicity of the drug. Baicalein also inhibits vascular endothelial growth factor (VEGF) expression etc. Analyzing their advantages and disadvantages in treating OC provides a new perspective on the role of SB and its flavonoids in OC treatment. It serves as a resource for future OC research and development.
Collapse
Affiliation(s)
- Jiaying Cai
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhelin He
- Endoscopy Center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Xiaoyan Chen
- Endoscopy Center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Jian Wang
- Endoscopy Center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Xiang Yin
- Endoscopy Center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| |
Collapse
|
4
|
Czogalla B, Dötzer K, Sigrüner N, von Koch FE, Brambs CE, Anthuber S, Frangini S, Burges A, Werner J, Mahner S, Mayer B. Combined Expression of HGFR with Her2/neu, EGFR, IGF1R, Mucin-1 and Integrin α2β1 Is Associated with Aggressive Epithelial Ovarian Cancer. Biomedicines 2022; 10:2694. [PMID: 36359213 PMCID: PMC9687566 DOI: 10.3390/biomedicines10112694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/15/2022] [Accepted: 10/21/2022] [Indexed: 09/10/2023] Open
Abstract
Hepatocyte growth factor receptor (HGFR), also known as c-mesenchymal-epithelial transition factor (c-MET), plays a crucial role in the carcinogenesis of epithelial ovarian cancer (EOC). In contrast, the mechanisms contributing to aberrant expression of HGFR in EOC are not fully understood. In the present study, the expression of HGFR with its prognostic and predictive role was evaluated immunohistochemically in a cohort of 42 primary ovarian cancer patients. Furthermore, we analyzed the dual expression of HGFR and other druggable biomarkers. In the multivariate Cox regression analysis, high HGFR expression was identified as an independent prognostic factor for a shorter progression-free survival (PFS) (hazard ratio (HR) 2.99, 95% confidence interval (CI95%) 1.01-8.91, p = 0.049) and overall survival (OS) (HR 5.77, CI95% 1.56-21.34, p = 0.009). In addition, the combined expression of HGFR, human epidermal growth factor receptor 2 (Her2/neu), epithelial growth factor receptor (EGFR), insulin-like growth factor 1 (IGF1R), Mucin-1 and Integrin α2β1 further significantly impaired PFS, platinum-free interval (PFI) and OS. Protein co-expression analyses were confirmed by transcriptomic data in a large, independent cohort of patients. In conclusion, new biomarker-directed treatment targets were identified to fight poor prognosis of primary EOC.
Collapse
Affiliation(s)
- Bastian Czogalla
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University Munich, Marchioninistraße 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Pettenkoferstraße 8a, 80336 Munich, Germany
| | - Katharina Dötzer
- Department of General, Visceral and Transplant Surgery, University Hospital, Ludwig-Maximilians-University Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Nicole Sigrüner
- Department of General, Visceral and Transplant Surgery, University Hospital, Ludwig-Maximilians-University Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Franz Edler von Koch
- Gynecology and Obstetrics Clinic, Klinikum Dritter Orden, Menzinger Straße 44, 80638 Munich, Germany
| | - Christine E. Brambs
- Department of Obstetrics and Gynecology, Klinikum Rechts der Isar, Technical University Munich, Ismaninger Straße 22, 81675 Munich, Germany
| | - Sabine Anthuber
- Department of Obstetrics and Gynecology, Starnberg Hospital, Oßwaldstraße 1, 82319 Starnberg, Germany
| | - Sergio Frangini
- Department of Obstetrics and Gynecology, Munich Clinic Harlaching, Sanatoriumsplatz 2, 81545 Munich, Germany
| | - Alexander Burges
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University Munich, Marchioninistraße 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Pettenkoferstraße 8a, 80336 Munich, Germany
| | - Jens Werner
- German Cancer Consortium (DKTK), Partner Site Munich, Pettenkoferstraße 8a, 80336 Munich, Germany
- Department of General, Visceral and Transplant Surgery, University Hospital, Ludwig-Maximilians-University Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University Munich, Marchioninistraße 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Pettenkoferstraße 8a, 80336 Munich, Germany
| | - Barbara Mayer
- German Cancer Consortium (DKTK), Partner Site Munich, Pettenkoferstraße 8a, 80336 Munich, Germany
- Department of General, Visceral and Transplant Surgery, University Hospital, Ludwig-Maximilians-University Munich, Marchioninistraße 15, 81377 Munich, Germany
| |
Collapse
|
5
|
Design, synthesis and evaluation of calix[4]arene-based carbonyl amide derivatives with antitumor activities. Eur J Med Chem 2020; 210:112984. [PMID: 33183867 DOI: 10.1016/j.ejmech.2020.112984] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/18/2020] [Accepted: 10/30/2020] [Indexed: 11/23/2022]
Abstract
Calixarenes, with potential functionalization on the upper and lower rim, have been explored in recent years for the design and construction of anticancer agents in the field of drugs and pharmaceuticals. Herein, optimization of bis [N-(2-hydroxyethyl) aminocarbonylmethoxyl substituted calix [4] arene (CLX-4) using structure-based drug design and traditional medicinal chemistry led to the discovery of series of calix [4]arene carbonyl amide derivatives 5a-5t. Evaluation of the cytotoxicity of 5a-5t employing MTT assay in MCF-7, MDA-MB-231 (human breast cancer cells), HT29 (human colon carcinoma cells), HepG2 (human hepatocellular carcinoma cells), A549 (human lung adenocarcinoma cells) and HUVEC (Human Umbilical Vein Endothelial) cells demonstrated that the most promising compound 5h displayed the most superior inhibitory effect against A549 and MDA-MB-231 cells, which were 3.2 times and 6.8 times of CLX-4, respectively. In addition, the cell inhibition rate (at 10 μM) against normal HUVEC cells in vitro was only 9.6%, indicating the safty of compound 5h. Moreover, compound 5h could inhibit the migration of MDA-MB-231 cell in wound healing assay. Further mechanism studies significantly indicated that compound 5h could block MDA-MB-231 cell cycle arrest in G0/G1 phase by down regulating cyclin D1 and CDK4, and induce apoptosis by up-regulation of Bax, down-regulation of Caspase-3, PARP and Bcl-2 proteins, resulting in the reduction of DNA synthesis and cell division arrest. This work provides worthy of further exploration for the promising calixarene-based anticancer drugs.
Collapse
|
6
|
Delayed recanalization after MCAO ameliorates ischemic stroke by inhibiting apoptosis via HGF/c-Met/STAT3/Bcl-2 pathway in rats. Exp Neurol 2020; 330:113359. [PMID: 32428505 DOI: 10.1016/j.expneurol.2020.113359] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/28/2020] [Accepted: 05/11/2020] [Indexed: 01/28/2023]
Abstract
The activation of tyrosine kinase receptor c-Met by hepatocyte growth factor (HGF) showed an anti-apoptotic effect in numerous disease models. This study aimed to investigate the neuroprotective mechanism of the HGF/c-Met axis-mediated anti-apoptosis underlying the delayed recanalization in a rat model of middle cerebral artery occlusion (MCAO). Permanent MCAO model (pMCAO) was induced by intravascular filament insertion. Recanalization was induced by withdrawing the filament at 3 days after MCAO (rMCAO). HGF levels in the blood serum and brain tissue expressions of HGF, c-Met, phosphorylated-STAT3 (p-STAT3), STAT3, Bcl-2, Bax, cleaved caspase-3(CC3) were assessed using ELISA and western blot, respectively. To study the mechanism, HGF small interfering ribonucleic acid (siRNA) and c-Met inhibitor, su11274, were administered intracerebroventricularly (i.c.v.) or intranasally, respectively. The concentration of HGF in the serum was increased significantly after MCAO. Brain expression of HGF was increased after MCAO and peaked at 3 days after recanalization. HGF and c-Met were both co-localized with neurons. Compared to rats received permanent MCAO, delayed recanalization after MCAO decreased the infarction volume, inhibited neuronal apoptosis, and improved neurobehavioral function, increased expressions of p-STAT3 and its downstream Bcl-2. Mechanistic studies indicated that HGF siRNA and su11274 reversed the neuroprotection including anti-apoptotic effects provided by delayed recanalization. In conclusion, the delayed recanalization after MCAO increased the expression of HGF in the brain, and reduced the infarction and neuronal apoptosis after MCAO, partly via the activation of the HGF/c-Met/STAT3/Bcl-2 signaling pathway. The delayed recanalization may serve as a therapeutic alternative for a subset of ischemic stroke patients.
Collapse
|
7
|
Zhou H, Liu Q, Shi X, Liu Y, Cao D, Yang J. Distinct gene expression profiles associated with clinical outcomes in patients with ovarian clear cell carcinoma and high-grade serous ovarian carcinoma. J Ovarian Res 2020; 13:38. [PMID: 32295618 PMCID: PMC7161165 DOI: 10.1186/s13048-020-00641-8] [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: 09/24/2019] [Accepted: 03/31/2020] [Indexed: 12/28/2022] Open
Abstract
Background Ovarian clear cell carcinoma (OCCC) is the second most common ovarian cancer after serous carcinoma in Southeast Asia. OCCC has a more unfavourable clinical outcome due to a poor response to platinum-based chemotherapy compared with serous carcinoma. The identification of biomarkers related to the prognosis of OCCC is critically important for an improved understanding of the biology that drives OCCC progression and leads to poor outcomes. To detect differences in gene expression profiles between OCCC and high-grade serous ovarian carcinoma (HGSOC), twelve patients with OCCC and twelve patients with HGSOC were recruited in whom the pathological diagnosis was confirmed on surgically resected specimens. Results Compared with HGSOC, OCCC has 609 differentially expression genes, and 199 are significantly different (P < 0.05). These genes are involved in the cell cycle, apoptosis, DNA damage repair, the PI3K pathway and so on. There were 164 differentially expressed genes in the PI3K pathway. There were 35 overexpressed genes in OCCC, while there were 12 overexpressed genes in HGSOC. Among these differentially expressed genes, we found that the MET gene and the CCNE1 gene were overexpressed in OCCC and associated with a worse prognosis. Conclusions In conclusion, there are many differentially expressed genes in OCCC and HGSOC, which indicates that the two kinds of tumours differ greatly in tumourigenesis and provides a theoretical basis for targeted therapy in the future. Further studies need to be performed to clarify the association of the differentially expressed genes with the unfavourable prognosis in OCCC.
Collapse
Affiliation(s)
- Huimei Zhou
- Department of Gynaecologic Oncology, Peking Union Medical College Hospital, 1 Shuaifuyuan, Dongcheng-qu, Beijing, People's Republic of China
| | - Qian Liu
- Department of Gynaecologic Oncology, Peking Union Medical College Hospital, 1 Shuaifuyuan, Dongcheng-qu, Beijing, People's Republic of China
| | - Xiaohua Shi
- Department of Pathology, Peking Union Medical College Hospital, Beijing, People's Republic of China
| | - Yue Liu
- Department of Gynaecologic Oncology, Peking Union Medical College Hospital, 1 Shuaifuyuan, Dongcheng-qu, Beijing, People's Republic of China
| | - Dongyan Cao
- Department of Gynaecologic Oncology, Peking Union Medical College Hospital, 1 Shuaifuyuan, Dongcheng-qu, Beijing, People's Republic of China
| | - Jiaxin Yang
- Department of Gynaecologic Oncology, Peking Union Medical College Hospital, 1 Shuaifuyuan, Dongcheng-qu, Beijing, People's Republic of China.
| |
Collapse
|
8
|
Binju M, Amaya-Padilla MA, Wan G, Gunosewoyo H, Suryo Rahmanto Y, Yu Y. Therapeutic Inducers of Apoptosis in Ovarian Cancer. Cancers (Basel) 2019; 11:E1786. [PMID: 31766284 PMCID: PMC6896143 DOI: 10.3390/cancers11111786] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/06/2019] [Accepted: 11/06/2019] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancers remain one of the most common causes of gynecologic cancer-related death in women worldwide. The standard treatment comprises platinum-based chemotherapy, and most tumors develop resistance to therapeutic drugs. One mechanism of developing drug resistance is alterations of molecules involved in apoptosis, ultimately assisting in the cells' capability to evade death. Thus, there is a need to focus on identifying potential drugs that restore apoptosis in cancer cells. Here, we discuss the major inducers of apoptosis mediated through various mechanisms and their usefulness as potential future treatment options for ovarian cancer. Broadly, they can target the apoptotic pathways directly or affect apoptosis indirectly through major cancer-pathways in cells. The direct apoptotic targets include the Bcl-2 family of proteins and the inhibitor of apoptotic proteins (IAPs). However, indirect targets include processes related to homologous recombination DNA repair, micro-RNA, and p53 mutation. Besides, apoptosis inducers may also disturb major pathways converging into apoptotic signals including janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3), wingless-related integration site (Wnt)/β-Catenin, mesenchymal-epithelial transition factor (MET)/hepatocyte growth factor (HGF), mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK), and phosphatidylinositol 3-kinase (PI3K)/v-AKT murine thymoma viral oncogene homologue (AKT)/mammalian target of rapamycin (mTOR) pathways. Several drugs in our review are undergoing clinical trials, for example, birinapant, DEBIO-1143, Alisertib, and other small molecules are in preclinical investigations showing promising results in combination with chemotherapy. Molecules that exhibit better efficacy in the treatment of chemo-resistant cancer cells are of interest but require more extensive preclinical and clinical evaluation.
Collapse
Affiliation(s)
- Mudra Binju
- School of Pharmacy & Biomedical Sciences, Curtin University, Curtin Health Innovative Research Institute, Perth, WA 6102, Australia
| | - Monica Angelica Amaya-Padilla
- School of Pharmacy & Biomedical Sciences, Curtin University, Curtin Health Innovative Research Institute, Perth, WA 6102, Australia
| | - Graeme Wan
- School of Pharmacy & Biomedical Sciences, Curtin University, Curtin Health Innovative Research Institute, Perth, WA 6102, Australia
| | - Hendra Gunosewoyo
- School of Pharmacy & Biomedical Sciences, Curtin University, Curtin Health Innovative Research Institute, Perth, WA 6102, Australia
| | - Yohan Suryo Rahmanto
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| | - Yu Yu
- School of Pharmacy & Biomedical Sciences, Curtin University, Curtin Health Innovative Research Institute, Perth, WA 6102, Australia
- University of Western Australia Medical School, Division of Obstetrics & Gynaecology, Perth, WA 6009, Australia
| |
Collapse
|
9
|
Ko EJ, Oh YL, Kim HY, Eo WK, Kim H, Ock MS, Kim HS, Kim KH, Cha HJ. Correlation of long interspersed element-1 open reading frame 1 and c-Met proto-oncogene protein expression in ovarian cancer. Genes Genomics 2019; 41:1293-1299. [PMID: 31388980 DOI: 10.1007/s13258-019-00858-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hypomethylation of long interspersed nuclear element-1 (LINE-1) is closely related to certain cancers and concerns with aggressive tumor behavior. Previously, we reported LINE-1 open reading frame-1 (ORF1) protein level was significantly up-regulated in ovarian cancers compared with normal ovary. Hypomethylation of local LINE-1 sequence has been reported to reactivate MET proto-oncogene in colon cancers and hepatocellular carcinoma. However, the relationship between LINE-1 and c-MET expressions in ovarian cancer is not yet studied. METHOD Here, we analyzed the expression patterns of LINE-1 ORF1 and c-Met protein in ovarian cancer tissue microarrays containing 208 surgical specimens including normal ovary and malignant ovarian cancers. RESULTS The expressions of both LINE-1 ORF1 and c-Met protein were significantly increased in ovarian cancers and peaked in early stage of tumor. Other clinical data including age and tumor types were not significantly related with both proteins. Co-relationship between LINE-1 ORF1 and c-Met protein was significant (p = 0.03) but several patients show different expression patterns. CONCLUSIONS These results propose that LINE-1 ORF1 significantly activates c-Met but not in all cases, suggesting other factors may be involved simultaneously.
Collapse
Affiliation(s)
- Eun-Ji Ko
- Departments of Parasitology and Genetics, Kosin University College of Medicine, Busan, Republic of Korea.,Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
| | - Young Lim Oh
- Departments of Obstetrics and Gynecology, Kosin University College of Medicine, Busan, Republic of Korea
| | - Heung Yeol Kim
- Departments of Obstetrics and Gynecology, Kosin University College of Medicine, Busan, Republic of Korea
| | - Wan Kyu Eo
- Department of Internal Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hongbae Kim
- Department of Obstetrics and Gynecology, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Mee Sun Ock
- Departments of Parasitology and Genetics, Kosin University College of Medicine, Busan, Republic of Korea
| | - Heui-Soo Kim
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
| | - Ki Hyung Kim
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Busan, Republic of Korea. .,Biomedical Research Institute and Pusan Cancer Center, Pusan National University Hospital, Busan, Republic of Korea.
| | - Hee-Jae Cha
- Departments of Parasitology and Genetics, Kosin University College of Medicine, Busan, Republic of Korea.
| |
Collapse
|
10
|
Clinical significance of c-Met and phospho-c-Met (Tyr1234/1235) in ovarian cancer. Taiwan J Obstet Gynecol 2019; 58:105-110. [PMID: 30638462 DOI: 10.1016/j.tjog.2018.11.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2018] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE c-Met is expressed in human ovarian cancer tissues, and its phosphorylation activates signaling cascades that might affect the behavior of cancer cells. In this study, we evaluated the association of c-Met and phosphorylated c-Met (phospho-c-Met) expressions with the clinical outcomes of ovarian cancer patients. MATERIALS AND METHODS Archived tissue from surgical specimens of 269 ovarian cancer patients who underwent a debulking operation in MacKay Memorial Hospital between 2004 and 2012 were collected. Tissue microarrays were stained with anti-Met and anti-phospho-Met (Tyr1234/1235) monoclonal antibodies. Immunostaining intensity was scored on a scale of 0-3+. High expression was defined as more than 50% of moderate and intense staining. Patients' clinical data were reviewed until April 2017 for analysis. RESULTS The proportion of high c-Met expression was significantly higher in patients with cancer in early stages (Federation of Gynecology and Obstetrics stages I and II) and low histologic grades (grades 1 and 2) (79.70%, p = 0.0008 and 80.15%, p ≤ 0.0001, respectively). However, no association was found between phospho-c-Met and FIGO stage or the histologic grade. Ovarian clear cell carcinoma and mucinous carcinoma had much higher c-Met expression (95.16% and 87.10%, p ≤ 0.0001 and p = 0.0292, respectively). Although the overall survival did not differ significantly, low expressions of c-Met and phospho-c-Met were obviously associated with poor progression-free survival respectively (p = 0.0034, HR: 0.5264, 95% CI: 0.3326-0.8330 and p = 0.0136, HR: 0.5626, 95% CI: 0.3709-0.8535). CONCLUSION Low c-Met expression was associated with poor clinical outcomes.
Collapse
|
11
|
MacMahon P, Labak CM, Martin-Bach SE, Issawi A, Velpula K, Tsung AJ. Glioblastoma formation in a recurrent intracranial epidermoid cyst: a case report. CNS Oncol 2018; 7:CNS25. [PMID: 30543115 PMCID: PMC6331695 DOI: 10.2217/cns-2018-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background: Transformation to glioblastoma following recurrent epidermoid cyst resection has not been reported. Chronic inflammation can underlie malignant transformation of epidermoid cysts. Astrogliosis following repeated resections may have induced the rare transformation to glioblastoma. Clinical presentation: A patient presenting with left lower extremity weakness was found to harbor a parietal mass lesion. Histopathology demonstrated an epidermoid cyst. Following multiple re-resections, an intra-axial mass was discovered within the operative bed, confirmed as glioblastoma. Conclusion: This is the first report of glioblastoma associated with a resected epidermoid cyst. Subsequent to resection, the chronic inflammatory milieu propagated by astrogliosis is thought to have induced malignancy. The progression to glioblastoma draws attention to neoplastic transformation in the context of recurrent epidermoids.
Collapse
Affiliation(s)
- Paul MacMahon
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, 61605, USA.,Neurosurgery Department, OSF HealthCare Illinois Neurological Institute, Peoria, IL, 61637, USA
| | - Collin M Labak
- Department of Cancer Biology & Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, 61605, USA
| | - Sarah E Martin-Bach
- Department of Pathology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA.,Neuropathology, OSF HealthCare Illinois Neurological Institute, Peoria, IL, 61637, USA
| | - Ahmad Issawi
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, 61605, USA.,Neurosurgery Department, OSF HealthCare Illinois Neurological Institute, Peoria, IL, 61637, USA
| | - Kiran Velpula
- Department of Cancer Biology & Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, 61605, USA
| | - Andrew J Tsung
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, 61605, USA.,Neurosurgery Department, OSF HealthCare Illinois Neurological Institute, Peoria, IL, 61637, USA.,Department of Cancer Biology & Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, 61605, USA
| |
Collapse
|
12
|
Yu T, Ma P, Wu D, Shu Y, Gao W. Functions and mechanisms of microRNA-31 in human cancers. Biomed Pharmacother 2018; 108:1162-1169. [PMID: 30372817 DOI: 10.1016/j.biopha.2018.09.132] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs can exhibit opposite functions in different tumors. MiR-31 is a representative example as it can not only enhance tumor development and progression in pancreatic cancer, colorectal cancer and so on, but also inhibit tumorigenesis and induce apoptosis in ovarian cancer, prostate cancer and etc. The mechanism underlying its' pleiotropy remains unknown. Several recent studies that focused on the global gene expression changes caused by aberrant miR-31 provided information on the upstream and downstream events associated with deregulated miR-31. MiR-31 might interact with a number of signaling pathways including RAS/MARK, PI3K/AKT and RB/E2F to play its opposite functions. This review summarizes the target genes and pathways associated with miR-31 and examines the mechanisms underlying the function of miR-31. The resulting hypothesis is possible that the tissue-specific features of adenocarcinoma and squamous cell cancer and the positive feedback loop consists of miR-31 and its upstream and downstream may account for the diversity of miR-31 functions.
Collapse
Affiliation(s)
- Tao Yu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Pei Ma
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Deqin Wu
- Department of Pharmacy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Wen Gao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
13
|
Achkar IW, Abdulrahman N, Al-Sulaiti H, Joseph JM, Uddin S, Mraiche F. Cisplatin based therapy: the role of the mitogen activated protein kinase signaling pathway. J Transl Med 2018; 16:96. [PMID: 29642900 PMCID: PMC5896132 DOI: 10.1186/s12967-018-1471-1] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/02/2018] [Indexed: 12/19/2022] Open
Abstract
Cisplatin is a widely used chemotherapeutic agent for treatment of various cancers. However, treatment with cisplatin is associated with drug resistance and several adverse side effects such as nephrotoxicity, reduced immunity towards infections and hearing loss. A Combination of cisplatin with other drugs is an approach to overcome drug resistance and reduce toxicity. The combination therapy also results in increased sensitivity of cisplatin towards cancer cells. The mitogen activated protein kinase (MAPK) pathway in the cell, consisting of extracellular signal regulated kinase, c-Jun N-terminal kinase, p38 kinases, and downstream mediator p90 ribosomal s6 kinase (RSK); is responsible for the regulation of various cellular events including cell survival, cell proliferation, cell cycle progression, cell migration and protein translation. This review article demonstrates the role of MAPK pathway in cisplatin based therapy, illustrates different combination therapy involving cisplatin and also shows the importance of targeting MAPK family, particularly RSK, to achieve increased anticancer effect and overcome drug resistance when combined with cisplatin.
Collapse
Affiliation(s)
- Iman W. Achkar
- Translational Research Institute, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | | | - Hend Al-Sulaiti
- College of Pharmacy, Qatar University, P.O. Box 2713, Doha, Qatar
| | | | - Shahab Uddin
- Translational Research Institute, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Fatima Mraiche
- College of Pharmacy, Qatar University, P.O. Box 2713, Doha, Qatar
| |
Collapse
|
14
|
Ding C, Zou Q, Wang F, Wu H, Chen R, Lv J, Ling M, Sun J, Wang W, Li H, Huang B. Human amniotic mesenchymal stem cells improve ovarian function in natural aging through secreting hepatocyte growth factor and epidermal growth factor. Stem Cell Res Ther 2018. [PMID: 29523193 PMCID: PMC5845161 DOI: 10.1186/s13287-018-0781-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Although many reports show that various kinds of stem cells have the ability to recover function in premature ovarian aging, few studies have looked at stem cell treatment of natural ovarian aging (NOA). We designed this experimental study to investigate whether human amniotic mesenchymal stem cells (hAMSCs) retain the ability to restore ovarian function, and how hAMSCs work in this process. Methods To build the NOA mouse model, the mice were fed for 12–14 months normally with young fertile female mice as the normal control group (3–5 months old). Hematoxylin and eosin staining permitted follicle counting and showed the ovarian tissue structure. An enzyme-linked immunosorbent assay was used to detect the serum levels of the sex hormones estradiol (E2), anti-mullerian hormone (AMH), and follicle-stimulating hormone (FSH). The proliferation rate and marker expression level of human ovarian granule cells (hGCs) (ki67, AMH, FSH receptor, FOXL2, and CYP19A1) were measured by flow cytometry (FACS). Cytokines (growth factors) were measured by a protein antibody array methodology. After hepatocyte growth factor (HGF) and epidermal growth factor (EGF) were co-cultured with hGCs, proliferation (ki67) and apoptosis (Annexin V) levels were analyzed by FACS. After HGF and EGF were injected into the ovaries of natural aging mice, the total follicle numbers and hormone levels were tested. Results After the hAMSCs were transplanted into the NOA mouse model, the hAMSCs exerted a therapeutic activity on mouse ovarian function by improving the follicle numbers over four stages. In addition, our results showed that hAMSCs significantly promoted the proliferation rate and marker expression level of ovarian granular cells that were from NOA patients. Meanwhile, we found that the secretion level of EGF and HGF from hAMSCs was higher than other growth factors. A growth factor combination (HGF with EGF) improved the proliferation rate and inhibited the apoptosis rate more powerfully after a co-culture with hGCs, and total follicle numbers and hormone levels were elevated to a normal level after the growth factor combination was injected into the ovaries of the NOA mouse model. Conclusions These findings provide insight into the notion that hAMSCs play an integral role in resistance to NOA. Furthermore, our present study demonstrates that a growth factor combination derived from hAMSCs plays a central role in inhibiting ovarian aging. Therefore, we suggest that hAMSCs improve ovarian function in natural aging by secreting HGF and EGF. Electronic supplementary material The online version of this article (10.1186/s13287-018-0781-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chenyue Ding
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Qinyan Zou
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Fuxin Wang
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Huihua Wu
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Rulei Chen
- Central Laboratory, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Jinghuan Lv
- Central Laboratory, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Mingfa Ling
- Central Laboratory, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Jian Sun
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Wei Wang
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Hong Li
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China.
| | - Boxian Huang
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China. .,Central Laboratory, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China. .,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
15
|
Moran-Jones K. The Therapeutic Potential of Targeting the HGF/cMET Axis in Ovarian Cancer. Mol Diagn Ther 2017; 20:199-212. [PMID: 27139908 DOI: 10.1007/s40291-016-0201-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Survival rates for ovarian cancer have remained relatively stable for the past 2 decades despite advances in surgical techniques and cytotoxic chemotherapeutics, indicating a requirement for better therapies. One pathway currently proposed for targeting is the HGF/cMET pathway. Upregulated in a number of tumour types, cMET is a tyrosine kinase receptor expressed on epithelial cells. In ovarian cancer, it has been identified as highly expressed in the four major subtypes, with expression estimates ranging from 11 to 68 % of cases. HGF, the only known ligand for cMET, is found at high levels in both serum and ascites in women with ovarian cancer, and is proposed to induce both migration and metastasis. However, clinically validated biomarkers are not yet available for either HGF or cMET, preventing a clear understanding of the true rate of overexpression, or its correlation with prognosis. Despite this, a number of agents against HGF and cMET are currently being investigated in clinical trials for multiple tumour types, including ovarian. However, a lack of patient selection, biomarker usage, and post hoc analysis correlating response with expression has resulted in the majority of these trials showing little beneficial effect from these agents, indicating that additional research is required to determine their usefulness in patients with ovarian cancer.
Collapse
Affiliation(s)
- Kim Moran-Jones
- Wolfson Wohl Cancer Research Centre, University of Glasgow, Switchback Rd, Glasgow, G61 1QH, UK. .,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 370 Victoria St, Sydney, NSW, 2010, Australia.
| |
Collapse
|
16
|
Kim HJ, Yoon A, Ryu JY, Cho YJ, Choi JJ, Song SY, Bang H, Lee JS, Cho WC, Choi CH, Lee JW, Kim BG, Bae DS. c-MET as a Potential Therapeutic Target in Ovarian Clear Cell Carcinoma. Sci Rep 2016; 6:38502. [PMID: 27917934 PMCID: PMC5137074 DOI: 10.1038/srep38502] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 11/10/2016] [Indexed: 12/30/2022] Open
Abstract
In this study, we investigated the therapeutic effects of c-MET inhibition in ovarian clear cell carcinoma (OCCC). Expression levels of c-MET in the epithelial ovarian cancers (EOCs) and normal ovarian tissues were evaluated using real-time PCR. To test the effects of c-MET inhibitors in OCCC cell lines, we performed MTT and apoptosis assays. We used Western blots to evaluate the expression of c-MET and its down-stream pathway. In vivo experiments were performed to test the effects of c-MET inhibitor on tumor growth in orthotopic mouse xenografts of OCCC cell line RMG1 and a patient-derived tumor xenograft (PDX) model of OCCC. c-MET expression was significantly greater in OCCCs compared with serous carcinomas and normal ovarian tissues (p < 0.001). In in vitro study, inhibition of c-MET using c-MET inhibitors (SU11274 or crizotinib) significantly decreased the proliferation, and increased the apoptosis of OCCC cells. SU11274 decreased expression of the p-c-MET proteins and blocked the phosphorylation of down-stream proteins Akt and Erk. Furthermore, SU11274 treatment significantly decreased the in vivo tumor weight in xenograft models of RMG1 cell and a PDX model for OCCC compared to control (p = 0.004 and p = 0.009, respectively).
Collapse
Affiliation(s)
- Ha-Jeong Kim
- Department of Obstetrics and Gynecology, Institute of Wonkwang Medical Science, College of Medicine, Wonkwang University, Iksan, Korea
| | - Aera Yoon
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji-Yoon Ryu
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young-Jae Cho
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jung-Joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Yong Song
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Heejin Bang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Soo Lee
- Health promotion center Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - William Chi Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Chel Hun Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Byoung-Gie Kim
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Duk-Soo Bae
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
17
|
Kwon Y, Godwin AK. Regulation of HGF and c-MET Interaction in Normal Ovary and Ovarian Cancer. Reprod Sci 2016; 24:494-501. [PMID: 27170665 DOI: 10.1177/1933719116648212] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Binding of hepatocyte growth factor (HGF) to the c-MET receptor has mitogenic, motogenic, and morphogenic effects on cells. The versatile biological effects of HGF and c-MET interactions make them important contributors to the development of malignant tumors. We and others have demonstrated a therapeutic value in targeting the interaction of c-MET and HGF in epithelial ovarian cancer (EOC). However, both HGF and c-MET are expressed in the normal ovary as well. Therefore, it is important to understand the differences in mechanisms that control HGF signaling activation and its functional role in the normal ovary and EOC. In the normal ovary, HGF signaling may be under hormonal regulation. During ovulation, HGF-converting proteases are secreted and the subsequent activation of HGF signaling enhances the proliferation of ovarian surface epithelium in order to replenish the area damaged due to expulsion of the ovum. In contrast, EOC cells that exhibit epithelial characteristics constitutively express both c-MET and HGF-converting proteases such as urokinase-type plasminogen activator. In EOC, mechanisms to control the activation of HGF signaling are absent since HGF is provided locally from the tissue microenvironment as well as remotely throughout the body. Potential incessant HGF signaling in EOC may lead to an increase in proliferation, invasion through the stroma, and migration to other tissues of cancer cells. Therefore, targeting the interaction of c-MET and HGF would be beneficial in treating EOC.
Collapse
Affiliation(s)
- Youngjoo Kwon
- 1 Department of Food Science and Engineering, Ewha Womans University, Seoul, Korea
| | - Andrew K Godwin
- 2 Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA.,3 University of Kansas Cancer Center, Kansas City, KS, USA
| |
Collapse
|
18
|
Moran-Jones K, Brown LM, Samimi G. INC280, an orally available small molecule inhibitor of c-MET, reduces migration and adhesion in ovarian cancer cell models. Sci Rep 2015; 5:11749. [PMID: 26138303 PMCID: PMC5155610 DOI: 10.1038/srep11749] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 06/04/2015] [Indexed: 12/16/2022] Open
Abstract
5-year survival rates for ovarian cancer are approximately 40%, and for women diagnosed at late stage (the majority), just 27%. This indicates a dire need for new treatments to improve survival rates. Recent molecular characterization has greatly improved our understanding of the disease and allowed the identification of potential new targets. One such pathway of interest is the HGF/c-MET axis. Activation of the HGF/c-MET axis has been demonstrated in certain ovarian tumours, and been found to be associated with decreased overall survival, suggesting its potential as a therapeutic target. The objective of this study was to determine the efficacy of a novel, highly potent, orally-bioavailable c-MET inhibitor, INC280, in blocking cell phenotypes important in ovarian cancer metastasis. Using in vitro and ex vivo models, we demonstrate that INC280 inhibits HGF-induced c-MET, and reduces downstream signalling. HGF-stimulated chemotactic and random migration are decreased by INC280 treatment, to levels seen in non-stimulated cells. Additionally, HGF-induced adhesion of cancer cells to peritoneal tissue is significantly decreased by INC280 treatment. Overall, these data indicate that INC280 inhibits many cell behaviours that promote ovarian cancer metastasis, and merits further investigation as a therapeutic candidate in the treatment of patients with ovarian cancer.
Collapse
Affiliation(s)
- Kim Moran-Jones
- 1] Kinghorn Cancer Centre and Garvan Institute of Medical Research, Darlinghurst, NSW, Australia [2] St. Vincent's Clinical School, Faculty of Medicine, UNSW Australia, NSW, Australia
| | - Laura M Brown
- Kinghorn Cancer Centre and Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Goli Samimi
- 1] Kinghorn Cancer Centre and Garvan Institute of Medical Research, Darlinghurst, NSW, Australia [2] St. Vincent's Clinical School, Faculty of Medicine, UNSW Australia, NSW, Australia
| |
Collapse
|
19
|
Chaparro RE, Izutsu M, Sasaki T, Sheng H, Zheng Y, Sadeghian H, Qin T, von Bornstadt D, Herisson F, Duan B, Li JS, Jiang K, Pearlstein M, Pearlstein RD, Smith DE, Goldberg ID, Ayata C, Warner DS. Sustained functional improvement by hepatocyte growth factor-like small molecule BB3 after focal cerebral ischemia in rats and mice. J Cereb Blood Flow Metab 2015; 35:1044-53. [PMID: 25712497 PMCID: PMC4640251 DOI: 10.1038/jcbfm.2015.23] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 01/12/2015] [Accepted: 01/16/2015] [Indexed: 11/08/2022]
Abstract
Hepatocyte growth factor (HGF), efficacious in preclinical models of acute central nervous system injury, is burdened by administration of full-length proteins. A multiinstitutional consortium investigated the efficacy of BB3, a small molecule with HGF-like activity that crosses the blood-brain barrier in rodent focal ischemic stroke using Stroke Therapy Academic Industry Roundtable (STAIR) and Good Laboratory Practice guidelines. In rats, BB3, begun 6 hours after temporary middle cerebral artery occlusion (tMCAO) reperfusion, or permanent middle cerebral artery occlusion (pMCAO) onset, and continued for 14 days consistently improved long-term neurologic function independent of sex, age, or laboratory. BB3 had little effect on cerebral infarct size and no effect on blood pressure. BB3 increased HGF receptor c-Met phosphorylation and synaptophysin expression in penumbral tissue consistent with a neurorestorative mechanism from HGF-like activity. In mouse tMCAO, BB3 starting 10 minutes after reperfusion and continued for 14 days improved neurologic function that persisted for 8 weeks in some, but not all measures. Study in animals with comorbidities and those exposed to common stroke drugs are the next steps to complete preclinical assessment. These data, generated in independent, masked, and rigorously controlled settings, are the first to suggest that the HGF pathway can potentially be harnessed by BB3 for neurologic benefit after ischemic stroke.
Collapse
Affiliation(s)
- Rafael E Chaparro
- Department of Anesthesiology, Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, North Carolina, USA
| | - Miwa Izutsu
- Department of Anesthesiology, Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, North Carolina, USA
| | - Toshihiro Sasaki
- Department of Anesthesiology, Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, North Carolina, USA
| | - Huaxin Sheng
- Department of Anesthesiology, Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, North Carolina, USA
| | - Yi Zheng
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Homa Sadeghian
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Tao Qin
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Daniel von Bornstadt
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Fanny Herisson
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Bin Duan
- Angion Biomedica Corporation, Uniondale, New York, USA
| | - Jing-Song Li
- Angion Biomedica Corporation, Uniondale, New York, USA
| | - Kai Jiang
- Angion Biomedica Corporation, Uniondale, New York, USA
| | - Molly Pearlstein
- Department of Anesthesiology, Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, North Carolina, USA
| | - Robert D Pearlstein
- Department of Surgery, Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, North Carolina, USA
| | - David E Smith
- Angion Biomedica Corporation, Uniondale, New York, USA
| | | | - Cenk Ayata
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - David S Warner
- Department of Anesthesiology, Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, North Carolina, USA
- Department of Surgery, Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurobiology, Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
20
|
Jang SG, Yoo CW, Park SY, Kang S, Kim HK. Low expression of miR-449 in gynecologic clear cell carcinoma. Int J Gynecol Cancer 2014; 24:1558-63. [PMID: 25238166 DOI: 10.1097/igc.0000000000000267] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Clinical detection of ovarian clear cell carcinomas is important because of the poor prognosis. To identify microRNA profiles specific for clear cell carcinomas, microRNA expression profiles were compared between clear cell carcinomas and serous carcinomas of the ovary using microRNA microarray. In parallel, clear cell carcinomas were compared with germ cell tumors of the ovary. Six microRNAs differentially expressed between ovarian clear cell and serous carcinomas distinguished uterine clear cell carcinomas from endometrioid carcinomas. MiR-449 was underexpressed in both ovarian and uterine clear cell carcinomas. When germ cell tumors were compared with clear cell carcinomas of the ovary, miR-302d was the most significantly overexpressed microRNA in germ cell tumors. Thus, here we describe microRNA profiles characteristic for clear cell carcinomas of the ovary and uterus.
Collapse
MESH Headings
- Adenocarcinoma, Clear Cell/genetics
- Adenocarcinoma, Clear Cell/pathology
- Adenocarcinoma, Clear Cell/surgery
- Adolescent
- Adult
- Aged
- Biomarkers, Tumor/genetics
- Carcinoma, Endometrioid/genetics
- Carcinoma, Endometrioid/pathology
- Carcinoma, Endometrioid/surgery
- Child
- Cystadenocarcinoma, Serous/genetics
- Cystadenocarcinoma, Serous/pathology
- Cystadenocarcinoma, Serous/surgery
- Female
- Follow-Up Studies
- Humans
- MicroRNAs/genetics
- Middle Aged
- Neoplasm Grading
- Neoplasm Staging
- Neoplasms, Germ Cell and Embryonal/genetics
- Neoplasms, Germ Cell and Embryonal/pathology
- Neoplasms, Germ Cell and Embryonal/surgery
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/surgery
- Prognosis
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Young Adult
Collapse
Affiliation(s)
- Sang-Geun Jang
- National Cancer Center, Goyang, Gyeonggi, Republic of Korea
| | | | | | | | | |
Collapse
|
21
|
Sheng XJ, Li Z, Sun M, Wang ZH, Zhou DM, Li JQ, Zhao Q, Sun XF, Liu QC. MACC1 induces metastasis in ovarian carcinoma by upregulating hepatocyte growth factor receptor c-MET. Oncol Lett 2014; 8:891-897. [PMID: 25009663 PMCID: PMC4081430 DOI: 10.3892/ol.2014.2184] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 04/29/2014] [Indexed: 12/22/2022] Open
Abstract
Metastasis-associated in colon cancer 1 (MACC1) is a newly identified gene that has been shown to promote tumor cell invasion and metastasis. The present study investigated the effect of MACC1 downregulation on the biological characteristics of the ovarian cancer OVCAR3 cell line. In this study, MACC1 expression was blocked using the RNA interference technique. The downregulation of MACC1 mRNA and protein expression was confirmed using quantitative polymerase chain reaction and western blot analysis. The proliferative activity and adhesion rate of the cells were detected using cell counting kit-8 and a cell adhesion assay, while cell invasion was determined using a Matrigel invasion assay and migration capacity was observed using migration and wound-healing assays. A tube formation assay was also used to examine the angiogenic capacity of cells, and a luciferase assay was performed to assess whether MACC1 binds to the c-MET gene. The MACC1 mRNA and protein expression levels were significantly downregulated using sequence-specific small interfering RNA (siRNA). The inhibition of MACC1 expression markedly decreased the invasive, metastatic and angiogenic capacities of the cells, but only slightly inhibited growth and adhesion. In addition, a putative MACC1-binding site was identified in the 3′-untranslated region of c-MET. MACC1-siRNA was also found to significantly reduce the expression of the c-MET protein and a luciferase reporter assay confirmed that c-MET was the target gene of MACC1. These results demonstrated that the attenuation of MACC1 suppresses cell invasion and migration and that MACC1 may regulate cell metastasis through targeting the expression of c-MET. Inhibition of the function of MACC1 may represent a new strategy for treating ovarian cancer.
Collapse
Affiliation(s)
- Xiu-Jie Sheng
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Zhen Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Man Sun
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Zhi-Hui Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Dong-Mei Zhou
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Jian-Qi Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Qin Zhao
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Xiao-Fang Sun
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, Guangdong 510150, P.R. China
| | - Qi-Cai Liu
- Experimental Medical Research Center of Guangzhou Medical University, Guangzhou, Guangdong 510182, P.R. China
| |
Collapse
|
22
|
X-chromosome-linked inhibitor of apoptosis as a key factor for chemoresistance in clear cell carcinoma of the ovary. Br J Cancer 2014; 110:2881-6. [PMID: 24853184 PMCID: PMC4056063 DOI: 10.1038/bjc.2014.255] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 04/10/2014] [Accepted: 04/14/2014] [Indexed: 12/11/2022] Open
Abstract
Background: X-chromosome-linked inhibitor of apoptosis (XIAP) is one of the anti-apoptotic proteins leading to chemoresistance in several cancers. The aim of this study is to evaluate the impact of XIAP expression upon ovarian clear cell carcinoma (CCC) that has a platinum-resistant phenotype. Methods: Tissue microarrays made from 90 CCC patients were analysed for immunohistochemical expression levels of XIAP, c-Met, p-Akt and Bcl-XL. In addition, CCC cell lines were evaluated whether XIAP silencing could modulate sensitivity to platinum agent in vitro. Results: High XIAP expression was observed in 30 (33%) of 90 CCC cases, and was associated with c-Met (<0.01) and Bcl-XL (<0.01) expression. Cases with high XIAP expression had lower response rate to primary platinum-based chemotherapy (10% vs 65%, P=0.02). In stages II–IV tumours, high XIAP expression was related with worse progression-free survival (PFS, P=0.02). Furthermore, high XIAP expression was identified as an independent worse prognostic factor for PFS and overall survival. Finally, downregulation of XIAP using XIAP-specific small interfering RNA increased sensitivity to cisplatin in human cancer cells derived from CCC. Conclusions: X-chromosome-linked inhibitor of apoptosis expression was correlated with chemoresistance of primary chemotherapy, and identified as a prognostic marker for CCC. X-chromosome-linked inhibitor of apoptosis could be a candidate for new therapeutic target in CCC.
Collapse
|
23
|
Gadducci A, Sergiampietri C, Lanfredini N, Guiggi I. Micro-RNAs and ovarian cancer: the state of art and perspectives of clinical research. Gynecol Endocrinol 2014; 30:266-71. [PMID: 24479883 DOI: 10.3109/09513590.2013.871525] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Dysregulation of microRNA (mi-RNA) expression plays a major role in the development and progression of most human malignancies. Members of the miR-200 family, miR-182, miR-214 and miR-221 are frequently up-regulated, whereas miR-100, let-7i, miR-199a, miR-125b, mir-145 and miR-335 are often down-regulated in ovarian cancer compared with normal ovarian tissue. Most mi-RNA signatures are overlapping in different tumor histotypes but some mi-RNAs seem to be histotype specific. For instance, the endometrioid type shares with the serous and clear cell types the up-regulation of miR-200 family members, but also presents over-expression of miR-21, miR-202 and miR-205. Clear cell carcinoma has a significantly higher expression of miR-30a and miR-30a*, whereas mucinous histotype has elevated levels of miR-192/194. In vitro and in vivo investigations have shown that several mi-RNAs can modulate the sensitivity of ovarian cancer to platinum and taxane, and clinical studies have suggested that mi-RNA profiling may predict the outcome of patients with this malignancy. Some mi-RNAs could be used as biomarkers to identify patients that might benefit from the addition of molecularly targeted agents (i.e. anti-angiogenic agents, MET inhibitors and poly(ADP-ribose) polymerase (PARP) inhibitors) to standard chemotherapy. Moreover, mi-RNAs could represent potential targets for the development of novel therapies.
Collapse
Affiliation(s)
- Angiolo Gadducci
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa , Pisa , Italy
| | | | | | | |
Collapse
|
24
|
Trovato M, Torre ML, Ragonese M, Simone A, Scarfì R, Barresi V, Giuffrè G, Benvenga S, Angileri FF, Tuccari G, Trimarchi F, Ruggeri RM, Cannavò S. HGF/c-met system targeting PI3K/AKT and STAT3/phosphorylated-STAT3 pathways in pituitary adenomas: an immunohistochemical characterization in view of targeted therapies. Endocrine 2013; 44:735-43. [PMID: 23576023 DOI: 10.1007/s12020-013-9950-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 04/03/2013] [Indexed: 12/21/2022]
Abstract
The ligand/receptor hepatocyte growth factor (HGF)/c-met signaling system promotes cellular growth and angiogenesis through PI3K/phosphor-Akt and STAT3/phosphor-STAT3 downstream effectors. In this study, we have evaluated the expression of molecules of the HGF/c-met pathway in pituitary adenomas (PA). The expression of HGF, c-met, PI3K (p85αsubunit) pAkt, STAT3, and pSTAT3 was analyzed by immunohistochemistry in an archival series of 30 PA (12 non-functioning and 18 functioning; 25 macroadenomas and 5 microadenomas). PAs expressed all six proteins in tumor epithelial cells. The proportion of c-met(+ve) cells was greater than HGF(+ve) cells (49 ± 19 vs 34 ± 17 %, P < 0.01), the pAkt(+ve) cells greater than PI3K(+ve) cells (39 ± 16.0 vs 1.3 ± 0.5 %, P < 0.001), and the STAT3(+ve) cells greater than active pSTAT3(+ve) cells (14 ± 8 vs 7 ± 6 %, P < 0.01). Furthermore, endothelial Akt immunostaining was detected on the vascular surface area of 17 PAs, in macroadenomas more frequently than in microadenomas (82 vs 18 %). The percentage of immunostained endothelial cells was greater in macro than in microadenomas (19 ± 7 and 7 ± 3 %; P < 0.05). In conclusion, HGF and c-met are widely expressed in PA, and correlate with pAkt expression. These data, together with the finding of pAkt immunostaining on microvascular areas related to tumor size, suggest a major role of the pAKT signaling in tumor growth and angiogenesis. There might be practical implications for the targeted therapy of PA.
Collapse
Affiliation(s)
- Maria Trovato
- Department of Human Pathology, Pad D 4 piano-AOU Policlinico "G. Martino", via Consolare Valeria, 1, 98125, Messina, Italy,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Mitamura T, Watari H, Wang L, Kanno H, Hassan MK, Miyazaki M, Katoh Y, Kimura T, Tanino M, Nishihara H, Tanaka S, Sakuragi N. Downregulation of miRNA-31 induces taxane resistance in ovarian cancer cells through increase of receptor tyrosine kinase MET. Oncogenesis 2013; 2:e40. [PMID: 23552883 PMCID: PMC3641356 DOI: 10.1038/oncsis.2013.3] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Ovarian cancer is one of the most aggressive female reproductive tract tumors. Paclitaxel (PTX) is widely used for the treatment of ovarian cancer. However, ovarian cancers often acquire chemotherapeutic resistance to this agent. We investigated the mechanism of chemoresistance by analysis of microRNAs using the ovarian cancer cell line KFr13 and its PTX-resistant derivative (KFr13Tx). We found that miR-31 was downregulated in KFr13Tx cells, and that re-introduction of miR31 re-sensitized them to PTX both in vitro and in vivo. miR-31 was found to bind to the 3′-UTR of mRNA of MET, and the decrease in MET correlated to higher sensitivity to PTX. Furthermore, co-treatment of KFr13Tx cells with MET inhibitors sensitized the tumor cells to PTX both in vitro and in vivo. In addition, lower levels of miR31 and higher expression of MET in human ovarian cancer specimens were significantly correlated with PTX chemoresistance and poor prognosis. This study demonstrated miR31-dependent regulation of MET for chemoresistance of ovarian cancer, raising the possibility that combination therapy with a MET inhibitor and PTX will increase PTX efficacy.
Collapse
Affiliation(s)
- T Mitamura
- 1] Department of Obstetrics and Gynecology, Hokkaido University Graduate School of Medicine, Sapporo, Japan [2] Department of Cancer Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Marchion DC, Bicaku E, Xiong Y, Bou Zgheib N, Al Sawah E, Stickles XB, Judson PL, Lopez AS, Cubitt CL, Gonzalez-Bosquet J, Wenham RM, Apte SM, Berglund A, Lancaster JM. A novel c-Met inhibitor, MK8033, synergizes with carboplatin plus paclitaxel to inhibit ovarian cancer cell growth. Oncol Rep 2013; 29:2011-8. [PMID: 23467907 PMCID: PMC4536335 DOI: 10.3892/or.2013.2329] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 01/29/2013] [Indexed: 12/26/2022] Open
Abstract
Elevated serum levels of hepatocyte growth factor (HGF) and high tumor expression of c-Met are both indicators of poor overall survival from ovarian cancer (OVCA). In the present study, we evaluated the role of the HGF signaling pathway in OVCA cell line chemoresistance and OVCA patient overall survival as well as the influence of HGF/c-Met signaling inhibition on the sensitivity of OVCA cells to combinational carboplatin plus paclitaxel therapy. The prevalence of the HGF receptor, c-Met, was determined by immunohistochemistry in primary OVCA samples (n=79) and OVCA cell lines (n=41). The influence of the c-Met-specific inhibitor MK8033 on OVCA cell sensitivity to combinations of carboplatin plus paclitaxel was examined in a subset of OVCA cells (n=8) by CellTiter-Blue cell viability assays. Correlation tests were used to identify genes associated with response to MK8033 and carboplatin plus paclitaxel. Identified genes were evaluated for influence on overall survival from OVCA using principal component analysis (PCA) modeling in an independent clinical OVCA dataset (n=218). Immunohistochemistry analysis indicated that 83% of OVCA cells and 92% of primary OVCA expressed the HGF receptor, c-Met. MK8033 exhibited significant anti-proliferative effects against a panel of human OVCA cell lines. Combination index values determined by the Chou-Talalay isobologram equation indicated synergistic activity in combinations of MK8033 and carboplatin plus paclitaxel. Pearson's correlation identified a 47-gene signature to be associated with MK8033-carboplatin plus paclitaxel response. PCA modeling indicated an association of this 47-gene response signature with overall survival from OVCA (P=0.013). These data indicate that HGF/c-Met pathway signaling may influence OVCA chemosensitivity and overall patient survival. Furthermore, HGF/c-Met inhibition by MK8033 represents a promising new therapeutic avenue to increase OVCA sensitivity to carboplatin plus paclitaxel.
Collapse
Affiliation(s)
- Douglas C Marchion
- Department of Women's Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Greenall SA, Gherardi E, Liu Z, Donoghue JF, Vitali AA, Li Q, Murphy R, Iamele L, Scott AM, Johns TG. Non-agonistic bivalent antibodies that promote c-MET degradation and inhibit tumor growth and others specific for tumor related c-MET. PLoS One 2012; 7:e34658. [PMID: 22511956 PMCID: PMC3325269 DOI: 10.1371/journal.pone.0034658] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 03/06/2012] [Indexed: 11/18/2022] Open
Abstract
The c-MET receptor has a function in many human cancers and is a proven therapeutic target. Generating antagonistic or therapeutic monoclonal antibodies (mAbs) targeting c-MET has been difficult because bivalent, intact anti-Met antibodies frequently display agonistic activity, necessitating the use of monovalent antibody fragments for therapy. By using a novel strategy that included immunizing with cells expressing c-MET, we obtained a range of mAbs. These c-MET mAbs were tested for binding specificity and anti-tumor activity using a range of cell-based techniques and in silico modeling. The LMH 80 antibody bound an epitope, contained in the small cysteine-rich domain of c-MET (amino acids 519–561), that was preferentially exposed on the c-MET precursor. Since the c-MET precursor is only expressed on the surface of cancer cells and not normal cells, this antibody is potentially tumor specific. An interesting subset of our antibodies displayed profound activities on c-MET internalization and degradation. LMH 87, an antibody binding the loop connecting strands 3d and 4a of the 7-bladed β-propeller domain of c-MET, displayed no intrinsic agonistic activity but promoted receptor internalization and degradation. LMH 87 inhibited HGF/SF-induced migration of SK-OV-3 ovarian carcinoma cells, the proliferation of A549 lung cancer cells and the growth of human U87MG glioma cells in a mouse xenograft model. These results indicate that c-MET antibodies targeting epitopes controlling receptor internalization and degradation provide new ways of controlling c-MET expression and activity and may enable the therapeutic targeting of c-MET by intact, bivalent antibodies.
Collapse
Affiliation(s)
- Sameer A. Greenall
- Oncogenic Signaling Laboratory, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | | | - Zhanqi Liu
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research, Heidelberg, Victoria, Australia
| | - Jacqueline F. Donoghue
- Oncogenic Signaling Laboratory, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Angela A. Vitali
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research, Heidelberg, Victoria, Australia
| | - Qian Li
- Oncogenic Signaling Laboratory, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Roger Murphy
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research, Heidelberg, Victoria, Australia
| | - Luisa Iamele
- Medical Research Council Centre, Cambridge, United Kingdom
| | - Andrew M. Scott
- Tumour Targeting Laboratory, Ludwig Institute for Cancer Research, Heidelberg, Victoria, Australia
| | - Terrance G. Johns
- Oncogenic Signaling Laboratory, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
- * E-mail:
| |
Collapse
|
28
|
Bu R, Uddin S, Ahmed M, Hussain AR, Alsobhi S, Amin T, Al-Nuaim A, Al-Dayel F, Abubaker J, Bavi P, Al-Kuraya KS. c-Met inhibitor synergizes with tumor necrosis factor-related apoptosis-induced ligand to induce papillary thyroid carcinoma cell death. Mol Med 2012; 18:167-177. [PMID: 22113498 PMCID: PMC3320136 DOI: 10.2119/molmed.2011.00238] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 11/15/2011] [Indexed: 11/06/2022] Open
Abstract
The Met receptor tyrosine kinase is overexpressed and/or activated in variety of human malignancies. Previously we have shown that c-Met is overexpressed in Middle Eastern papillary thyroid carcinoma (PTC) and significantly associated with an aggressive phenotype, but its role has not been fully elucidated in PTC. The aim of this study was to determine the functional link between the c-Met/AKT signaling pathway and death receptor 5 (DR5) in a large cohort of PTC in a tissue microarray format followed by functional studies using PTC cell lines and nude mice. Our data showed that high expressions of p-Met and DR5 were significantly associated with an aggressive phenotype of PTC and correlated with BRAF mutation. Treatment of PTC cell lines with PHA665752, an inhibitor of c-Met tyrosine kinase, inhibited cell proliferation and induced apoptosis via the mitochondrial pathway in PTC cell lines. PHA665752 treatment or expression of c-Met small interfering (si)RNA resulted in dephosphorylation of c-Met, AKT and its downstream effector molecules. Furthermore, PHA665752 treatment upregulated DR5 expression via generation of reactive oxygen species in PTC cell lines, and synergistically potentiated death receptor-induced apoptosis with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Finally, cotreatment with PHA665752 and TRAIL caused more pronounced effects on PTC xenograft tumor growth in nude mice. Our data suggest that the c-Met/AKT pathway may be a potential target for therapeutic intervention for treatment of PTC refractory to conventionally therapeutic modalities.
Collapse
Affiliation(s)
- Rong Bu
- Human Cancer Genomic Research, Research Center, Riyadh, Saudi Arabia
| | - Shahab Uddin
- Human Cancer Genomic Research, Research Center, Riyadh, Saudi Arabia
| | - Maqbool Ahmed
- Human Cancer Genomic Research, Research Center, Riyadh, Saudi Arabia
| | - Azhar R Hussain
- Human Cancer Genomic Research, Research Center, Riyadh, Saudi Arabia
| | - Saif Alsobhi
- Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Tarek Amin
- Department of Endocrinology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Abdurahman Al-Nuaim
- Department of Endocrinology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fouad Al-Dayel
- Department of Pathology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Jehad Abubaker
- Human Cancer Genomic Research, Research Center, Riyadh, Saudi Arabia
| | - Prashant Bavi
- Human Cancer Genomic Research, Research Center, Riyadh, Saudi Arabia
| | | |
Collapse
|
29
|
c-MET protects breast cancer cells from apoptosis induced by sodium butyrate. PLoS One 2012; 7:e30143. [PMID: 22253909 PMCID: PMC3257283 DOI: 10.1371/journal.pone.0030143] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2011] [Accepted: 12/13/2011] [Indexed: 11/24/2022] Open
Abstract
Sodium Butyrate (NaBu) is regarded as a potential reagent for cancer therapy. In this study, a specific breast cancer cell population that is resistant NaBu treatment was identified. These cells possess cancer stem cell characters, such as the capability of sphere formation in vitro and high tumor incident rate (85%) in mouse model. Forty percent of the NaBu resistant cells express the cancer stem cells marker, the CD133, whereas only 10% intact cells present the CD133 antigen. Furthermore, the endogenous expressing c-MET contributes to the survival of cancer stem cell population from the treatment of NaBu. The CD133+ group also presents a higher level of c-MET. A combination treatment of MET siRNA and NaBu efficiently prohibited the breast cancer progression, and the incident rate of the tumor decrease to 18%. This study may help to develop a new and alternative strategy for breast cancer therapy.
Collapse
|
30
|
Potentiation of anticancer drugs: effects of pentoxifylline on neoplastic cells. Int J Mol Sci 2011; 13:369-82. [PMID: 22312258 PMCID: PMC3269692 DOI: 10.3390/ijms13010369] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 12/07/2011] [Accepted: 12/13/2011] [Indexed: 01/19/2023] Open
Abstract
The drug efflux activity of P-glycoprotein (P-gp, a product of the mdr1 gene, ABCB1 member of ABC transporter family) represents a mechanism by which tumor cells escape death induced by chemotherapeutics. In this study, we investigated the mechanisms involved in the effects of pentoxifylline (PTX) on P-gp-mediated multidrug resistance (MDR) in mouse leukemia L1210/VCR cells. Parental sensitive mouse leukemia cells L1210, and multidrug-resistant cells, L1210/VCR, which are characterized by the overexpression of P-gp, were used as experimental models. The cells were exposed to 100 μmol/L PTX in the presence or absence of 1.2 μmol/L vincristine (VCR). Western blot analysis indicated a downregulation of P-gp protein expression when multidrug-resistant L1210/VCR cells were exposed to PTX. The effects of PTX on the sensitization of L1210/VCR cells to VCR correlate with the stimulation of apoptosis detected by Annexin V/propidium iodide apoptosis necrosis kit and proteolytic activation of both caspase-3 and caspase-9 monitored by Western blot analysis. Higher release of matrix metalloproteinases (MMPs), especially MMP-2, which could be attenuated by PTX, was found in L1210/VCR than in L1210 cells by gelatin zymography in electrophoretic gel. Exposure of resistant cells to PTX increased the content of phosphorylated Akt kinase. In contrast, the presence of VCR eliminated the effects of PTX on Akt kinase phosphorylation. Taken together, we conclude that PTX induces the sensitization of multidrug-resistant cells to VCR via downregulation of P-gp, stimulation of apoptosis and reduction of MMPs released from drug-resistant L1210/VCR cells. These facts bring new insights into the mechanisms of PTX action on cancer cells.
Collapse
|
31
|
Wang L, Mezencev R, Bowen NJ, Matyunina LV, McDonald JF. Isolation and characterization of stem-like cells from a human ovarian cancer cell line. Mol Cell Biochem 2011; 363:257-68. [PMID: 22160925 DOI: 10.1007/s11010-011-1178-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 11/24/2011] [Indexed: 12/18/2022]
Abstract
Increasing evidence supports the existence of a subpopulation of cancer cells capable of self-renewal and differentiation into diverse cell lineages. These cancer stem-like or cancer-initiating cells (CICs) also demonstrate resistance to chemo- and radiotherapy and may function as a primary source of cancer recurrence. We report here on the isolation and in vitro propagation of multicellular ovarian cancer spheroids from a well-established ovarian cancer cell line (OVCAR-3). The spheroid-derived cells (SDCs) display self-renewal potential, the ability to produce differentiated progeny, and increased expression of genes previously associated with CICs. SDCs also demonstrate higher invasiveness, migration potential, and enhanced resistance to standard anticancer agents relative to parental OVCAR-3 cells. Furthermore, SDCs display up-regulation of genes associated with epithelial-to-mesenchymal transition (EMT), anticancer drug resistance and/or decreased susceptibility to apoptosis, as well as, down-regulation of genes typically associated with the epithelial cell phenotype and pro-apoptotic genes. Pathway and biological process enrichment analyses indicate significant differences between the SDCs and precursor OVCAR-3 cells in TGF-beta-dependent induction of EMT, regulation of lipid metabolism, NOTCH and Hedgehog signaling. Collectively, our results indicate that these SDCs will be a useful model for the study of ovarian CICs and for the development of novel CIC-targeted therapies.
Collapse
Affiliation(s)
- Lijuan Wang
- Ovarian Cancer Institute and School of Biology, Georgia Institute of Technology, 315 Ferst Dr, Atlanta, GA 30332-0363, USA
| | | | | | | | | |
Collapse
|
32
|
Uddin S, Hussain AR, Ahmed M, Al-Sanea N, Abduljabbar A, Ashari LH, Alhomoud S, Al-Dayel F, Bavi P, Al-Kuraya KS. Coexpression of activated c-Met and death receptor 5 predicts better survival in colorectal carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:3032-3044. [PMID: 21978492 PMCID: PMC3260832 DOI: 10.1016/j.ajpath.2011.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2011] [Revised: 08/03/2011] [Accepted: 08/09/2011] [Indexed: 02/04/2023]
Abstract
Dysregulated overexpression of hepatocyte growth factor and its receptor, c-Met, has been reported in various cancers, but its role in colorectal carcinoma (CRC) has not been elucidated. Therefore, we investigated the role of phosphorylated Met (p-Met) in Middle Eastern CRC patient samples and cell lines. The p-Met was overexpressed in 80.8% of CRCs and strongly associated with the expression of p-AKT, DR5, and Ki-67 by immunohistochemistry. Coexpression of p-Met and DR5 was seen in 53.1% of CRC cases and was associated with a less aggressive phenotype, characterized by a histological subtype of adenocarcinomas, well-differentiated tumors, and was an independent prognostic marker for better overall survival. PHA665752, a selective p-Met inhibitor, induced apoptosis in CRC cells via inactivation of c-Met and AKT. PHA665752 treatment also caused increased expression of DR5 via generation of reactive oxygen species, and combination treatment with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and PHA665752 induced significant apoptosis. In vivo, cotreatment of a CRC xenograft with PHA665752 and TRAIL significantly reduced tumor volume and weight. These data demonstrate a significant correlation between p-Met and DR5 in patients with CRC. Furthermore, inhibition of p-Met signaling by PHA665752 in combination with TRAIL significantly inhibited cell growth and induced apoptosis in CRC cell lines, suggesting that this may have significant clinical implications as a therapeutic target in the treatment of CRC.
Collapse
Affiliation(s)
- Shahab Uddin
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Azhar R. Hussain
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Maqbool Ahmed
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nasser Al-Sanea
- Colorectal Unit, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Alaa Abduljabbar
- Colorectal Unit, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Luai H. Ashari
- Colorectal Unit, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Samar Alhomoud
- Colorectal Unit, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fouad Al-Dayel
- Department of Pathology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Prashant Bavi
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Khawla S. Al-Kuraya
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
33
|
Aune G, Lian AM, Tingulstad S, Torp SH, Forsmo S, Reseland JE, Stunes AK, Syversen U. Increased circulating hepatocyte growth factor (HGF): a marker of epithelial ovarian cancer and an indicator of poor prognosis. Gynecol Oncol 2011; 121:402-6. [PMID: 21284996 DOI: 10.1016/j.ygyno.2010.12.355] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 12/20/2010] [Accepted: 12/22/2010] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Hepatocyte growth factor (HGF) has been described to be increased in different cancers. In the present study we wanted to investigate whether HGF in serum can distinguish between benign and malignant ovarian tumors, and whether serum HGF levels can predict the outcome in patients with ovarian carcinomas. METHODS We included 123 consecutive patients appointed for laparotomy due to a pelvic mass. Preoperative levels of serum cancer antigen 125 (CA 125), HGF and HGF activator (HGFA) were quantified with immunological methods. We performed immunohistochemical analyses of HGFα, HGFβ and the receptor c-Met. Five-year survival of patients with advanced disease (stage III and stage IV) was analyzed with the Kaplan-Meier method. RESULTS Sixty patients had ovarian carcinomas, 23 borderline tumors, and 40 benign ovarian tumors. Patients with ovarian carcinomas had significantly higher preoperative HGF and CA 125 serum levels than patients with benign ovarian tumors, and borderline tumors. Patients with borderline tumors had significantly higher CA 125 values than benign cases. A combination of CA 125 and HGF increased the specificity in predicting carcinoma. We observed abundant HGFα, HGFβ and c-Met expressions in all ovarian tumors. Patients with advanced disease and preoperative serum HGF values ≥2SD above reference value had a shorter disease-free survival than patients with advanced disease and serum HGF <2SD above reference value. CONCLUSIONS HGF in serum is an indicator of ovarian carcinoma in women with a pelvic mass, and of a poor prognosis in advanced ovarian cancer.
Collapse
Affiliation(s)
- Guro Aune
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | | | | | | | | | | | | | | |
Collapse
|