1
|
Chang CS, Bai LY, Chiu CF, Hu JL, Weng JR. Discovery of the tryptanthrin-derived indoloquinazoline as an anti-breast cancer agent via ERK/JNK activation. ENVIRONMENTAL TOXICOLOGY 2024; 39:3710-3720. [PMID: 38511855 DOI: 10.1002/tox.24226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/25/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024]
Abstract
Tryptanthrin, an alkaloid applied in traditional Chinese medicine, exhibits a variety of pharmacological activities. This study aimed to investigate the anti-tumor activity of the tryptanthrin derivative (8-cyanoindolo[2,1-b]quinazoline-6,12-dione [CIQ]) in breast cancer cells. In both MDA-MB-231 and MCF-7 breast cancer cells, CIQ inhibited cell viability and promoted caspase-dependent apoptosis. At the concentration- and time-dependent ways, CIQ increased the levels of p-ERK, p-JNK, and p-p38 in breast cancer cells. We found that exposure to the JNK inhibitor or the ERK inhibitor partially reversed CIQ's viability. We also observed that CIQ increased reactive oxygen species (ROS) generation, and upregulated the phosphorylation and expression of H2AX. However, the pretreatment of the antioxidants did not protect the cells against CIQ's effects on cell viability and apoptosis, which suggested that ROS does not play a major role in the mechanism of action of CIQ. In addition, CIQ inhibited the invasion of MDA-MB-231 cells and decreased the expression of the prometastatic factors (MMP-2 and Snail). These findings demonstrated that the possibility of this compound to show promise in playing an important role against breast cancer.
Collapse
Affiliation(s)
| | - Li-Yuan Bai
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Chang-Fang Chiu
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- Cancer Center, China Medical University Hospital, Taichung, Taiwan
| | - Jing-Lan Hu
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Jing-Ru Weng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Master Degree Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
2
|
Dowaidar M. Guidelines for the role of autophagy in drug delivery vectors uptake pathways. Heliyon 2024; 10:e30238. [PMID: 38707383 PMCID: PMC11066435 DOI: 10.1016/j.heliyon.2024.e30238] [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: 01/25/2024] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024] Open
Abstract
The process of autophagy refers to the intracellular absorption of cytoplasm (such as proteins, nucleic acids, tiny molecules, complete organelles, and so on) into the lysosome, followed by the breakdown of that cytoplasm. The majority of cellular proteins are degraded by a process called autophagy, which is both a naturally occurring activity and one that may be induced by cellular stress. Autophagy is a system that can save cells' integrity in stressful situations by restoring metabolic basics and getting rid of subcellular junk. This happens as a component of an endurance response. This mechanism may have an effect on disease, in addition to its contribution to the homeostasis of individual cells and tissues as well as the control of development in higher species. The main aim of this study is to discuss the guidelines for the role of autophagy in drug delivery vector uptake pathways. In this paper, we discuss the meaning and concept of autophagy, the mechanism of autophagy, the role of autophagy in drug delivery vectors, autophagy-modulating drugs, nanostructures for delivery systems of autophagy modulators, etc. Later in this paper, we talk about how to deliver chemotherapeutics, siRNA, and autophagy inducers and inhibitors. We also talk about how hard it is to make a drug delivery system that takes nanocarriers' roles as autophagy modulators into account.
Collapse
Affiliation(s)
- Moataz Dowaidar
- Bioengineering Department, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia
- Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia
- Biosystems and Machines Research Center, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia
| |
Collapse
|
3
|
Wang J, Li B, Luo M, Huang J, Zhang K, Zheng S, Zhang S, Zhou J. Progression from ductal carcinoma in situ to invasive breast cancer: molecular features and clinical significance. Signal Transduct Target Ther 2024; 9:83. [PMID: 38570490 PMCID: PMC10991592 DOI: 10.1038/s41392-024-01779-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 04/05/2024] Open
Abstract
Ductal carcinoma in situ (DCIS) represents pre-invasive breast carcinoma. In untreated cases, 25-60% DCIS progress to invasive ductal carcinoma (IDC). The challenge lies in distinguishing between non-progressive and progressive DCIS, often resulting in over- or under-treatment in many cases. With increasing screen-detected DCIS in these years, the nature of DCIS has aroused worldwide attention. A deeper understanding of the biological nature of DCIS and the molecular journey of the DCIS-IDC transition is crucial for more effective clinical management. Here, we reviewed the key signaling pathways in breast cancer that may contribute to DCIS initiation and progression. We also explored the molecular features of DCIS and IDC, shedding light on the progression of DCIS through both inherent changes within tumor cells and alterations in the tumor microenvironment. In addition, valuable research tools utilized in studying DCIS including preclinical models and newer advanced technologies such as single-cell sequencing, spatial transcriptomics and artificial intelligence, have been systematically summarized. Further, we thoroughly discussed the clinical advancements in DCIS and IDC, including prognostic biomarkers and clinical managements, with the aim of facilitating more personalized treatment strategies in the future. Research on DCIS has already yielded significant insights into breast carcinogenesis and will continue to pave the way for practical clinical applications.
Collapse
Affiliation(s)
- Jing Wang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Baizhou Li
- Department of Pathology, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Meng Luo
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
- Department of Plastic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Huang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Kun Zhang
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu Zheng
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Suzhan Zhang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
| | - Jiaojiao Zhou
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
4
|
Oria RS, Anyanwu GE, Nto JN, Ikpa JO. Curcumin abrogates cobalt-induced neuroinflammation by suppressing proinflammatory cytokines release, inhibiting microgliosis and modulation of ERK/MAPK signaling pathway. J Chem Neuroanat 2024; 137:102402. [PMID: 38428651 DOI: 10.1016/j.jchemneu.2024.102402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/03/2024]
Abstract
Curcumin, a bioactive polyphenol derived from turmeric, has been reported to have anti-inflammatory properties. The current study investigated the anti-inflammatory effect of curcumin in the hippocampal subfields (CA1 and CA3) after exposure to cobalt (Co) and the impact of ERK protein. Twenty-eight albino Wistar rats were divided into four groups, each with seven randomly selected rats as follows: Control (distilled water), Cobalt (Co) only (40 mg/kg), 120 mg/kg or 240 mg/kg curcumin + Co (40 mg/kg). Treatment was via oral gavage for 28 days. We performed a biochemical investigation to determine the levels of proinflammatory cytokines (TNFα and IL-1β). Furthermore, we conducted an immunohistochemical evaluation to assess the expression of IBA1 by microglial cells and the immunoexpression of ERK protein in the hippocampus. Results revealed a significant (p<0.05) elevation in the tissue level of TNFα and IL-1β, an increase in the number of IBA1-positive microglia, and upregulation of ERK protein in the hippocampal subfields of the rats after exposure to cobalt-only. Nevertheless, pretreatment with curcumin restored these parameters to levels comparable to control. In conclusion, our results showed that curcumin abrogated the Co-induced neuroinflammation by suppressing the release of proinflammatory biomarkers, reducing microgliosis, and modulating the ERK/MAPK pathway.
Collapse
Affiliation(s)
- Rademene S Oria
- Department of Anatomy, Faculty Of Basic Medical Sciences, University of Cross River State (UNICROSS), Cross River State, Nigeria; Department Of Anatomy, Faculty Of Basic Medical Sciences, College Of Medicine, University Of Nigeria Enugu Campus,, Enugu, Nigeria.
| | - Godson E Anyanwu
- Department Of Anatomy, Faculty Of Basic Medical Sciences, College Of Medicine, University Of Nigeria Enugu Campus,, Enugu, Nigeria; Department of Anatomy, Faculty of Biomedical Sciences, Kampala International University, Uganda
| | - Johnson N Nto
- Department Of Anatomy, Faculty Of Basic Medical Sciences, College Of Medicine, University Of Nigeria Enugu Campus,, Enugu, Nigeria
| | - James O Ikpa
- Department of Anatomy, Faculty Of Basic Medical Sciences, University of Cross River State (UNICROSS), Cross River State, Nigeria
| |
Collapse
|
5
|
Zhou F, He K, Cai JJ, Davidson LA, Chapkin RS, Ni Y. A Unified Bayesian Framework for Bi-overlapping-Clustering Multi-omics Data via Sparse Matrix Factorization. STATISTICS IN BIOSCIENCES 2023; 15:669-691. [PMID: 38179127 PMCID: PMC10766378 DOI: 10.1007/s12561-022-09350-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 04/10/2021] [Accepted: 06/06/2022] [Indexed: 11/27/2022]
Abstract
The advances of modern sequencing techniques have generated an unprecedented amount of multi-omics data which provide great opportunities to quantitatively explore functional genomes from different but complementary perspectives. However, distinct modalities/sequencing technologies generate diverse types of data which greatly complicate statistical modeling because uniquely optimized methods are required for handling each type of data. In this paper, we propose a unified framework for Bayesian nonparametric matrix factorization that infers overlapping bi-clusters for multi-omics data. The proposed method adaptively discretizes different types of observations into common latent states on which cluster structures are built hierarchically. The proposed Bayesian nonparametric method is able to automatically determine the number of clusters. We demonstrate the utility of the proposed method using simulation studies and applications to a single-cell RNA-sequencing dataset, a combination of single-cell RNA-sequencing and single-cell ATAC-sequencing dataset, a bulk RNA-sequencing dataset, and a DNA methylation dataset which reveal several interesting findings that are consistent with biological literature.
Collapse
Affiliation(s)
- Fangting Zhou
- Institute of Statistics and Big Data, Renmin University of China, Beijing, China
- Department of Statistics, Texas A&M University, College Station, USA
| | - Kejun He
- Institute of Statistics and Big Data, Renmin University of China, Beijing, China
| | - James J. Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, USA
| | - Laurie A. Davidson
- Department of Nutrition and Food Science, Texas A&M University, College Station, USA
- Program in Integrative Nutrition and Complex Diseases, Texas A &M University, College Station, USA
| | - Robert S. Chapkin
- Department of Nutrition and Food Science, Texas A&M University, College Station, USA
- Program in Integrative Nutrition and Complex Diseases, Texas A &M University, College Station, USA
| | - Yang Ni
- Department of Statistics, Texas A&M University, College Station, USA
| |
Collapse
|
6
|
Zhang X, Liu J, Sun Y, Zhou Q, Ding X, Chen X. Chinese herbal compound Huangqin Qingrechubi capsule reduces lipid metabolism disorder and inflammatory response in gouty arthritis via the LncRNA H19/APN/PI3K/AKT cascade. PHARMACEUTICAL BIOLOGY 2023; 61:541-555. [PMID: 36994890 PMCID: PMC10064824 DOI: 10.1080/13880209.2023.2191641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/30/2023] [Accepted: 03/12/2023] [Indexed: 06/19/2023]
Abstract
CONTEXT Gouty arthritis (GA) is a characteristically inflammatory disease often associated with lipid metabolism disorder. Huangqin Qingrechubi capsule (HQC) has been used for the treatment of GA. OBJECTIVE To explore the mechanism of HQC in the treatment of GA. MATERIALS AND METHODS A total of 30 GA patients (GA group) and 30 healthy subjects [normal control (NC) group] were recruited. The GA group was treated with HQC (3.6 g/d) for 10 days. Lipid metabolism and inflammation indexes were detected. Five herbal names of HQC, or 'gouty arthritis', 'hyperlipidemia' and 'inflammation' were used as key words to search related databases for network pharmacological analysis. Subsequently, GA-fibroblast-like synoviocytes (FLSs) were stimulated with GA-peripheral blood mononuclear cells (PBMCs) (3:1) and treated with HQC drug-containing serum (20%). RT-qPCR, Western blot, and ELISA were conducted to further explore the mechanism of HQC in improving GA. RESULTS In clinical observation, HQC decreased the expression of lncRNA H19 and IL-1β, and increased the expression of adiponectin (APN) and IL-4 in the GA group (about half). Through network pharmacology, the PI3K/AKT signaling pathway was identified. Cell experiments showed that HQC treatment reduced the viability of GA-FLSs (49.61%), up-regulated the expression of IL-4 (155.18%), IL-10 (165.13%), and APN (31.24%), and down-regulated the expression of lncRNA H19 (33.70%), IL-1β (64.70%), TNF-α (78.32%), p-PI3K (48.80%), and p-AKT (53.48%). DISCUSSION AND CONCLUSIONS HQC improved lipid metabolism disorder and inflammatory response of GA by regulating the lncRNA H19/APN/PI3K/AKT. Maintaining the stability of lipid metabolism may be an effective way to alleviate GA.
Collapse
Affiliation(s)
- Xianheng Zhang
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui, University of Traditional Chinese Medicine, Hefei, China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, China
- Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Jian Liu
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui, University of Traditional Chinese Medicine, Hefei, China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, China
| | - Yanqiu Sun
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui, University of Traditional Chinese Medicine, Hefei, China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, China
- Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Qin Zhou
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui, University of Traditional Chinese Medicine, Hefei, China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, China
- Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Xiang Ding
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui, University of Traditional Chinese Medicine, Hefei, China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, China
- Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Xiaolu Chen
- Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui, University of Traditional Chinese Medicine, Hefei, China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, China
- Anhui University of Traditional Chinese Medicine, Hefei, China
| |
Collapse
|
7
|
Kuo SH, Wei MF, Lee YH, Lin JC, Yang WC, Yang SY, Huang CS. MAP3K1 expression is associated with progression and poor prognosis of hormone receptor-positive, HER2-negative early-stage breast cancer. Cell Oncol (Dordr) 2023; 46:1213-1234. [PMID: 37166744 DOI: 10.1007/s13402-023-00805-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2023] [Indexed: 05/12/2023] Open
Abstract
PURPOSE In this study, we assessed whether the overexpression of MAP3K1 promotes the proliferation, migration, and invasion of breast cancer cells, which affect the prognosis of hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative early stage breast cancer. METHODS Two HR-positive, HER2-negative breast cancer cell lines (MCF7 and T-47D) overexpressing MAP3K1 were transfected with two MAP3K1 short hairpin RNA plasmids (shMAP3K1 [#3] and shMAP3K1 [#5]). The proliferation, migration, and invasion of these cells were then examined. We assessed whether shMAP3K1 affects the cell cycle, levels of downstream signaling molecules (ERK, JNK, p38 MAPK, and NF-κB), and sensitivity to chemotherapeutic and hormonal agents. To assess the anti-tumor effect of MAP3K1 knockdown in the breast cancer orthotopic model, MCF7 and T-47D cells treated with or without shMAP3K1 (#3) and shMAP3K1 (#5) were inoculated into the mammary fat pads of mice. In total, 182 patients with HR-positive, HER2-negative T1 and T2 breast cancer and 0-3 nodal metastases were included. Additionally, 73 patients with T1 and T2 breast cancer and negative nodes who received adjuvant endocrine therapy alone were selected as an independent validation cohort. RESULTS In both cell lines, shMAP3K1 (#3) and shMAP3K1 (#5) significantly reduced cell growth, migration, and invasion by downregulating MMP-9 and by blocking the G2/M phase of the cell cycle and its regulatory molecule cyclin B1. Moreover, both shMAP3K1 (#3) and shMAP3K1 (#5) downregulated ERK-, JNK-, p38 MAPK-, and NF-κB-dependent gene transcription and enhanced the sensitivity of both cell lines to doxorubicin, docetaxel, and tamoxifen. We observed that both shMAP3K1 (#3) and shMAP3K1 (#5) inhibited tumor growth compared with that in the scrambled group of MCF7 and T-47D cell orthotopic tumors. Patients with MAP3K1 overexpression exhibited significantly poorer 10-year disease-free survival (DFS) (70.4% vs. 88.6%, p = 0.003) and overall survival (OS) (81.9% vs. 96.3%, p = 0.001) than those without MAP3K1 overexpression. Furthermore, phospho-ERK (p < 0.001) and phospho-JNK (p < 0.001) expressions were significantly associated with MAP3K1 expression, and both phospho-ERK and phospho-JNK expressions were significantly correlated with poor 10-year DFS and OS. These biological findings, including a significant association between DFS and OS, and the expressions of MAP3K1, phospho-ERK, and phospho-JNK were further validated in an independent cohort. Multivariate analysis identified MAP3K1 expression as an independent poor prognostic factor for DFS and OS. CONCLUSION Our results indicate that the overexpression of MAP3K1 plays a major role in the poor prognosis of HR-positive, HER2-negative early stage breast cancer.
Collapse
Affiliation(s)
- Sung-Hsin Kuo
- Departments of Oncology, National Taiwan University Hospital , Taipei, Taiwan
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Cancer Research Center, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Feng Wei
- Departments of Oncology, National Taiwan University Hospital , Taipei, Taiwan
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Cancer Research Center, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Departments of Pathology, National Taiwan University Hospital, and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jui-Chueh Lin
- Departments of Oncology, National Taiwan University Hospital , Taipei, Taiwan
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Cancer Research Center, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Chi Yang
- Departments of Oncology, National Taiwan University Hospital , Taipei, Taiwan
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Cancer Research Center, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shi-Yi Yang
- Department of Surgery, National Taiwan University Hospital, and College of Medicine, National Taiwan University, No. 7, Chung-Shan South Rd, Taipei, Taiwan
- Graduate Institute of Epidemiology, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chiun-Sheng Huang
- Department of Surgery, National Taiwan University Hospital, and College of Medicine, National Taiwan University, No. 7, Chung-Shan South Rd, Taipei, Taiwan.
| |
Collapse
|
8
|
Pasupuleti SK, Chao K, Ramdas B, Kanumuri R, Palam LR, Liu S, Wan J, Annesley C, Loh ML, Stieglitz E, Burke MJ, Kapur R. Potential clinical use of azacitidine and MEK inhibitor combination therapy in PTPN11-mutated juvenile myelomonocytic leukemia. Mol Ther 2023; 31:986-1001. [PMID: 36739480 PMCID: PMC10124140 DOI: 10.1016/j.ymthe.2023.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/17/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare myeloproliferative neoplasm of childhood. The molecular hallmark of JMML is hyperactivation of the Ras/MAPK pathway with the most common cause being mutations in the gene PTPN11, encoding the protein tyrosine phosphatase SHP2. Current strategies for treating JMML include using the hypomethylating agent, 5-azacitidine (5-Aza) or MEK inhibitors trametinib and PD0325901 (PD-901), but none of these are curative as monotherapy. Utilizing an Shp2E76K/+ murine model of JMML, we show that the combination of 5-Aza and PD-901 modulates several hematologic abnormalities often seen in JMML patients, in part by reducing the burden of leukemic hematopoietic stem and progenitor cells (HSC/Ps). The reduced JMML features in drug-treated mice were associated with a decrease in p-MEK and p-ERK levels in Shp2E76K/+ mice treated with the combination of 5-Aza and PD-901. RNA-sequencing analysis revealed a reduction in several RAS and MAPK signaling-related genes. Additionally, a decrease in the expression of genes associated with inflammation and myeloid leukemia was also observed in Shp2E76K/+ mice treated with the combination of the two drugs. Finally, we report two patients with JMML and PTPN11 mutations treated with 5-Aza, trametinib, and chemotherapy who experienced a clinical response because of the combination treatment.
Collapse
Affiliation(s)
- Santhosh Kumar Pasupuleti
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN 46202, USA
| | - Karen Chao
- Department of Pediatrics, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Stanford University School of Medicine, Lucile Packard Children's Hospital, Palo Alto, CA, USA
| | - Baskar Ramdas
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN 46202, USA
| | - Rahul Kanumuri
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN 46202, USA
| | - Lakshmi Reddy Palam
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN 46202, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | - Elliot Stieglitz
- Department of Pediatrics, Benioff Children's Hospital, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Michael J Burke
- Department of Pediatrics, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Reuben Kapur
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN 46202, USA; Department of Microbiology & Immunology, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN 46202, USA.
| |
Collapse
|
9
|
Weber LI, Hartl M. Strategies to target the cancer driver MYC in tumor cells. Front Oncol 2023; 13:1142111. [PMID: 36969025 PMCID: PMC10032378 DOI: 10.3389/fonc.2023.1142111] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/10/2023] [Indexed: 03/29/2023] Open
Abstract
The MYC oncoprotein functions as a master regulator of cellular transcription and executes non-transcriptional tasks relevant to DNA replication and cell cycle regulation, thereby interacting with multiple proteins. MYC is required for fundamental cellular processes triggering proliferation, growth, differentiation, or apoptosis and also represents a major cancer driver being aberrantly activated in most human tumors. Due to its non-enzymatic biochemical functions and largely unstructured surface, MYC has remained difficult for specific inhibitor compounds to directly address, and consequently, alternative approaches leading to indirect MYC inhibition have evolved. Nowadays, multiple organic compounds, nucleic acids, or peptides specifically interfering with MYC activities are in preclinical or early-stage clinical studies, but none of them have been approved so far for the pharmacological treatment of cancer patients. In addition, specific and efficient delivery technologies to deliver MYC-inhibiting agents into MYC-dependent tumor cells are just beginning to emerge. In this review, an overview of direct and indirect MYC-inhibiting agents and their modes of MYC inhibition is given. Furthermore, we summarize current possibilities to deliver appropriate drugs into cancer cells containing derailed MYC using viral vectors or appropriate nanoparticles. Finding the right formulation to target MYC-dependent cancers and to achieve a high intracellular concentration of compounds blocking or attenuating oncogenic MYC activities could be as important as the development of novel MYC-inhibiting principles.
Collapse
|
10
|
Waszczykowska K, Prażanowska K, Kałuzińska Ż, Kołat D, Płuciennik E. Discovering biomarkers for hormone-dependent tumors: in silico study on signaling pathways implicated in cell cycle and cytoskeleton regulation. Mol Genet Genomics 2022; 297:947-963. [PMID: 35532795 DOI: 10.1007/s00438-022-01900-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/16/2022] [Indexed: 02/07/2023]
Abstract
Malignancies dependent on hormone homeostasis include breast, ovary, cervical, prostate, testis and uterine tumors. Hormones are involved in signal transduction which orchestrate processes, such as apoptosis, proliferation, cell cycle or cytoskeleton organization. Currently, there is a need for novel biomarkers which would help to diagnose cancers efficiently. In this study, the genes implicated in signaling that is important in hormone-sensitive carcinogenesis were investigated regarding their prognostic significance. Data of seven cancer cohorts were collected from FireBrowse. 54 gene sets implicated in specific pathways were browsed through MSig database. Profiling was assessed via Monocle3, while gene ontology through PANTHER. For confirmation, correlation analysis was performed using WGCNA. Protein-protein networks were visualized via Cytoscape and impact of genes on survival, as well as cell cycle or cytoskeleton-related prognostic signatures, was tested. Several differences in expression profile were identified, some of them allowed to distinguish histology. Functional annotation revealed that various regulation of cell cycle, adhesion, migration, apoptosis and angiogenesis underlie these differences. Clinical traits, such as histological type or cancer staging, were found during evaluation of module-trait relationships. Of modules, the TopHubs (COL6A3, TNR, GTF2A1, NKX3-1) interacted directly with, e.g., PDGFB, ITGA10, SP1 or AKT3. Among TopHubs and interacting proteins, many showed an impact on hazard ratio and affected the cell cycle or cytoskeleton-related prognostic signatures, e.g., COL1A1 or PDGFB. In conclusion, this study laid the foundation for further hormone-sensitive carcinogenesis research through identification of genes which prove that crosstalk between cell cycle and cytoskeleton exists, opening avenues for future therapeutic strategies.
Collapse
Affiliation(s)
| | - Karolina Prażanowska
- Faculty of Biomedical Sciences, Medical University of Lodz, 90-752, Lodz, Poland
| | - Żaneta Kałuzińska
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland.
| | - Damian Kołat
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland
| | - Elżbieta Płuciennik
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland
| |
Collapse
|
11
|
Therapeutic potential of the PI3K inhibitor LY294002 and PARP inhibitor Talazoparib combination in BRCA-deficient triple negative breast cancer cells. Cell Signal 2021; 91:110229. [PMID: 34958867 DOI: 10.1016/j.cellsig.2021.110229] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/08/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023]
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors provide a promising therapeutic strategy for triple-negative breast cancers (TNBCs) with BRCA1/2 mutation. However, acquire resistance mechanisms and genetic alterations limit the clinical efficacy of PARP inhibitors. The aberrant activation of phosphatidylinositol 3-kinase (PI3K) is a significant problem for cancer development and thus the inhibition of PI3K by PI3K inhibitors is a novel targeted therapy in advanced breast cancer. Here, we, for the first time, investigated that the combined inhibition of PARP by Talazoparib (TAL) and PI3K by LY294002 synergistically inhibited proliferation of BRCA1 mutant HCC1937 TNBC cells through apoptosis, G0/G1 arrest, oxidative stress and increased DNA damage compared to drug alone. Additionally, TAL and LY294002 combination could be a promising strategy for overcoming TAL resistance. Co-treatment of TAL with LY294002 considerably suppressed the activation of PI3K, Akt1 and mTOR expression and phosphorylated protein levels in TNBC cells and caused changes in the multiple kinase phosphorylation. Our findings revealed that the dual inhibition of PARP and PI3K might represent an effective therapeutic strategy for TNBC and potentially overcome TAL resistance.
Collapse
|
12
|
Richard V, Davey MG, Annuk H, Miller N, Dwyer RM, Lowery A, Kerin MJ. MicroRNAs in Molecular Classification and Pathogenesis of Breast Tumors. Cancers (Basel) 2021; 13:5332. [PMID: 34771496 PMCID: PMC8582384 DOI: 10.3390/cancers13215332] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/21/2022] Open
Abstract
The current clinical practice of breast tumor classification relies on the routine immunohistochemistry-based expression analysis of hormone receptors, which is inadequate in addressing breast tumor heterogeneity and drug resistance. MicroRNA expression profiling in tumor tissue and in the circulation is an efficient alternative to intrinsic molecular subtyping that enables precise molecular classification of breast tumor variants, the prediction of tumor progression, risk stratification and also identifies critical regulators of the tumor microenvironment. This review integrates data from protein, gene and miRNA expression studies to elaborate on a unique miRNA-based 10-subtype taxonomy, which we propose as the current gold standard to allow appropriate classification and separation of breast cancer into a targetable strategy for therapy.
Collapse
Affiliation(s)
- Vinitha Richard
- Discipline of Surgery, The Lambe Institute for Translational Research, National University of Ireland, H91 YR71 Galway, Ireland; (M.G.D.); (H.A.); (N.M.); (R.M.D.); (A.L.)
| | | | | | | | | | | | - Michael J. Kerin
- Discipline of Surgery, The Lambe Institute for Translational Research, National University of Ireland, H91 YR71 Galway, Ireland; (M.G.D.); (H.A.); (N.M.); (R.M.D.); (A.L.)
| |
Collapse
|
13
|
Yu CY, Mitrofanova A. Mechanism-Centric Approaches for Biomarker Detection and Precision Therapeutics in Cancer. Front Genet 2021; 12:687813. [PMID: 34408770 PMCID: PMC8365516 DOI: 10.3389/fgene.2021.687813] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
Biomarker discovery is at the heart of personalized treatment planning and cancer precision therapeutics, encompassing disease classification and prognosis, prediction of treatment response, and therapeutic targeting. However, many biomarkers represent passenger rather than driver alterations, limiting their utilization as functional units for therapeutic targeting. We suggest that identification of driver biomarkers through mechanism-centric approaches, which take into account upstream and downstream regulatory mechanisms, is fundamental to the discovery of functionally meaningful markers. Here, we examine computational approaches that identify mechanism-centric biomarkers elucidated from gene co-expression networks, regulatory networks (e.g., transcriptional regulation), protein-protein interaction (PPI) networks, and molecular pathways. We discuss their objectives, advantages over gene-centric approaches, and known limitations. Future directions highlight the importance of input and model interpretability, method and data integration, and the role of recently introduced technological advantages, such as single-cell sequencing, which are central for effective biomarker discovery and time-cautious precision therapeutics.
Collapse
Affiliation(s)
- Christina Y. Yu
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Antonina Mitrofanova
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
14
|
Spirina LV, Avgustinovich AV, Afanas'ev SG, Cheremisina OV, Volkov MY, Choynzonov EL, Gorbunov AK, Usynin EA. Molecular Mechanism of Resistance to Chemotherapy in Gastric Cancers, the Role of Autophagy. Curr Drug Targets 2021; 21:713-721. [PMID: 31775598 DOI: 10.2174/1389450120666191127113854] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/11/2019] [Accepted: 11/21/2019] [Indexed: 12/13/2022]
Abstract
Gastric cancer (GC) is biologically and genetically heterogeneous with complex carcinogenesis at the molecular level. Despite the application of multiple approaches in the GC treatment, its 5-year survival is poor. A major limitation of anti-cancer drugs application is intrinsic or acquired resistance, especially to chemotherapeutical agents. It is known that the effectiveness of chemotherapy remains debatable and varies according to the molecular type of GC. Chemotherapy has an established role in the management of GC. Perioperative chemotherapy or postoperative chemotherapy is applied for localized ones. Most of the advanced GC patients have a poor response to treatment and unfavorable outcomes with standard therapies. Resistance substantially limits the depth and duration of clinical responses to targeted anticancer therapies. Through the use of complementary experimental approaches, investigators have revealed that cancer cells can achieve resistance through adaptation or selection driven by specific genetic, epigenetic, or microenvironmental alterations. Ultimately, these diverse alterations often lead to the activation of MAPK, AKT/mTOR, and Wnt/β-catenin signaling pathways that, when co-opted, enable cancer cells to survive drug treatments. We have summarized the mechanisms of resistance development to cisplatin, 5-fluorouracil, and multidrug resistance in the GC management. The complexity of molecular targets and components of signaling cascades altered in the resistance development results in the absence of significant benefits in GC treatment, and its efficacy remains low. The universal process responsible for the failure in the multimodal approach in GC treatment is autophagy. Its dual role in oncogenesis is the most unexplored issue. We have discussed the possible mechanism of autophagy regulation upon the action of endogenous factors and drugs. The experimental data obtained in the cultured GC cells need further verification. To overcome the cancer resistance and to prevent autophagy as the main reason of ineffective treatment, it is suggested the concept of the direct influence of autophagy molecular markers followed by the standard chemotherapy. Dozen of studies have focused on finding the rationale for the benefits of such complex therapy. The perspectives in the molecular-based management of GC are associated with the development of molecular markers predicting the protective autophagy initiation and search for novel targets of effective anticancer therapy.
Collapse
Affiliation(s)
- Liudmila V Spirina
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 5 Koopertivny street, Tomsk, 634050, Russian Federation.,Siberian State Medical University, 2, Moskovsky trakt, Tomsk, 634050, Russian Federation
| | - Alexandra V Avgustinovich
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 5 Koopertivny street, Tomsk, 634050, Russian Federation
| | - Sergey G Afanas'ev
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 5 Koopertivny street, Tomsk, 634050, Russian Federation
| | - Olga V Cheremisina
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 5 Koopertivny street, Tomsk, 634050, Russian Federation
| | - Maxim Yu Volkov
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 5 Koopertivny street, Tomsk, 634050, Russian Federation
| | - Evgeny L Choynzonov
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 5 Koopertivny street, Tomsk, 634050, Russian Federation.,Siberian State Medical University, 2, Moskovsky trakt, Tomsk, 634050, Russian Federation
| | - Alexey K Gorbunov
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 5 Koopertivny street, Tomsk, 634050, Russian Federation
| | - Evgeny A Usynin
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 5 Koopertivny street, Tomsk, 634050, Russian Federation.,Siberian State Medical University, 2, Moskovsky trakt, Tomsk, 634050, Russian Federation
| |
Collapse
|
15
|
Elghobashy YA, Assar MFA, Mahmoud AA, Monem A Eltorgoman A, Elmasry S. The relation between mitogen activated protein kinase (MAPK) pathway and different genes expression in patients with beta Thalassemia. Biochem Biophys Rep 2020; 24:100836. [PMID: 33195827 PMCID: PMC7644576 DOI: 10.1016/j.bbrep.2020.100836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND β-thalassemia is an inherited hemoglobinopathy resulting in quantitative changes in the β-globin chain. Understanding the molecular basis of that disorder requires studying the expression of genes controlling the pathways that affect the erythropoietic homeostasis especially the MAPK pathway. The MAPKs are a family of serine/threonine kinases that play an essential role in connecting cell-surface receptors to DNA in the nucleus of the cell. AIM to study the effect of expression of GNAI2, DUSP5 and ARRB1 genes on MAPK signaling pathway in pediatric patients with beta thalassemia. METHODS Forty children with beta thalassemia major (TM), forty children with beta thalassemia intermedia (TI) and forty age and gender matched healthy controls were enrolled in this study. Detection of GNAI2, DUSP5 and ARRB1 mRNA expression was done by real time polymerase chain reaction (RT-PCR). RESULTS revealed increased expression of ARRB1 (Arrestin Beta 1) gene, and decreased expression of both GNAI2 (Guanine nucleotide-binding protein G (i) subunit alpha-2) and DUSP5 (Dual specificity protein phosphatase 5) genes in both patient groups than control groups respectively. CONCLUSIONS Change in the rate of expression of ARRB1, GNAI2 and DUSP5 may have a role in the pathogenesis of abnormal hematopoiesis in cases of β thalassemia through affecting the MAPK pathway.
Collapse
Affiliation(s)
- Yasser AbdElsattar Elghobashy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
| | - Mohamed FA. Assar
- Biochemistry Division of Chemistry Department, Faculty of Science, Menoufia University, Shebin Elkom, Egypt
| | - Asmaa A. Mahmoud
- Department of Pediatrics, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
| | | | - Saher Elmasry
- Biochemistry Division of Chemistry Department, Faculty of Science, Menoufia University, Shebin Elkom, Egypt
| |
Collapse
|
16
|
Jung YJ, Park W, Kang KP, Kim W. SIRT2 is involved in cisplatin-induced acute kidney injury through regulation of mitogen-activated protein kinase phosphatase-1. Nephrol Dial Transplant 2020; 35:1145-1156. [PMID: 32240312 DOI: 10.1093/ndt/gfaa042] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 02/04/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Activation of mitogen-activated protein kinase phosphatase-1 (MKP-1), a dual-specificity protein phosphatase, regulates mitogen-activated protein kinase signaling. C-Jun N-terminal kinase (JNK) and p38 are activated in cisplatin-induced renal injury. However, the change of MKP-1 expression in cisplatin-induced renal injury and the regulatory effect of sirtuin 2 (SIRT2), a nicotinamide adenine dinucleotide-dependent deacetylase, on MKP-1 remains unknown. METHODS To address these issues, we used constitutional Sirt2 knockout (KO) mice, transgenic (TG) mice with increased expression of SIRT2 specifically in proximal tubular epithelial cellsand wild-type (WT) mice. Cisplatin nephrotoxicity was induced by intraperitoneal injection of cisplatin. RESULTS MKP-1 expression in the kidney was decreased after cisplatin treatment. Cisplatin-induced downregulation of MKP-1 was reversed in Sirt2 KO mice kidney and further decreased in Sirt2 TG mice kidney. We observed similar phenomenon with SIRT2-knockdown or SIRT2-overexpressed tubular epithelial cells. Phosphorylation of p38 and JNK, a downstream signal pathway of MKP-1, increased in WT mice kidney following treatment with cisplatin. A decrease in SIRT2 suppressed cisplatin-induced phosphorylation of p38 and JNK in kidney and tubular epithelial cells. Overexpression of SIRT2 further increased phosphorylation of p38 and JNK in kidney and tubular epithelial cells. Acetylation of MKP-1 was significantly increased in SIRT2-knockdown cells and decreased in SIRT2-overexpressed cells after cisplatin stimulation. Sirt2 KO mice and Sirt2 TG mice showed amelioration and aggravation of renal injury, apoptosis, necroptosis and inflammation induced by cisplatin. CONCLUSION Our data show that SIRT2 is associated with cisplatin-induced renal injury through regulation of MKP-1 expression.
Collapse
Affiliation(s)
- Yu Jin Jung
- Department of Internal Medicine, Division of Nephrology, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Woong Park
- Department of Internal Medicine, Division of Nephrology, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Kyung Pyo Kang
- Department of Internal Medicine, Division of Nephrology, Chonbuk National University Medical School, Jeonju, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Won Kim
- Department of Internal Medicine, Division of Nephrology, Chonbuk National University Medical School, Jeonju, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| |
Collapse
|
17
|
Lapcik P, Pospisilova A, Janacova L, Grell P, Fabian P, Bouchal P. How Different Are the Molecular Mechanisms of Nodal and Distant Metastasis in Luminal A Breast Cancer? Cancers (Basel) 2020; 12:E2638. [PMID: 32947901 PMCID: PMC7563588 DOI: 10.3390/cancers12092638] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 11/17/2022] Open
Abstract
Lymph node status is one of the best prognostic factors in breast cancer, however, its association with distant metastasis is not straightforward. Here we compare molecular mechanisms of nodal and distant metastasis in molecular subtypes of breast cancer, with major focus on luminal A patients. We analyze a new cohort of 706 patients (MMCI_706) as well as an independent cohort of 836 primary tumors with full gene expression information (SUPERTAM_HGU133A). We evaluate the risk of distant metastasis, analyze targetable molecular mechanisms in Gene Set Enrichment Analysis and identify relevant inhibitors. Lymph node positivity is generally associated with NF-κB and Src pathways and is related to high risk (OR: 5.062 and 2.401 in MMCI_706 and SUPERTAM_HGU133A, respectively, p < 0.05) of distant metastasis in luminal A patients. However, a part (≤15%) of lymph node negative tumors at the diagnosis develop the distant metastasis which is related to cell proliferation control and thrombolysis. Distant metastasis of lymph node positive patients is mostly associated with immune response. These pro-metastatic mechanisms further vary in other molecular subtypes. Our data indicate that the management of breast cancer and prevention of distant metastasis requires stratified approach based on targeted strategies.
Collapse
Affiliation(s)
- Petr Lapcik
- Department of Biochemistry, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic; (P.L.); (A.P.); (L.J.)
| | - Anna Pospisilova
- Department of Biochemistry, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic; (P.L.); (A.P.); (L.J.)
| | - Lucia Janacova
- Department of Biochemistry, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic; (P.L.); (A.P.); (L.J.)
| | - Peter Grell
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, 65653 Brno, Czech Republic;
| | - Pavel Fabian
- Department of Oncological Pathology, Masaryk Memorial Cancer Institute, 65653 Brno, Czech Republic;
| | - Pavel Bouchal
- Department of Biochemistry, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic; (P.L.); (A.P.); (L.J.)
| |
Collapse
|
18
|
Wen X, Ou YC, Bogatcheva G, Thomas G, Mahadevan-Jansen A, Singh B, Lin EC, Bardhan R. Probing metabolic alterations in breast cancer in response to molecular inhibitors with Raman spectroscopy and validated with mass spectrometry. Chem Sci 2020; 11:9863-9874. [PMID: 34094246 PMCID: PMC8162119 DOI: 10.1039/d0sc02221g] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/19/2020] [Indexed: 01/07/2023] Open
Abstract
Rapid and accurate response to targeted therapies is critical to differentiate tumors that are resistant to treatment early in the regimen. In this work, we demonstrate a rapid, noninvasive, and label-free approach to evaluate treatment response to molecular inhibitors in breast cancer (BC) cells with Raman spectroscopy (RS). Metabolic reprogramming in BC was probed with RS and multivariate analysis was applied to classify the cells into responsive or nonresponsive groups as a function of drug dosage, drug type, and cell type. Metabolites identified with RS were then validated with mass spectrometry (MS). We treated triple-negative BC cells with Trametinib, an inhibitor of the extracellular-signal-regulated kinase (ERK) pathway. Changes measured with both RS and MS corresponding to membrane phospholipids, amino acids, lipids and fatty acids indicated that these BC cells were responsive to treatment. Comparatively, minimal metabolic changes were observed post-treatment with Alpelisib, an inhibitor of the mammalian target of rapamycin (mTOR) pathway, indicating treatment resistance. These findings were corroborated with cell viability assay and immunoblotting. We also showed estrogen receptor-positive MCF-7 cells were nonresponsive to Trametinib with minimal metabolic and viability changes. Our findings support that oncometabolites identified with RS will ultimately enable rapid drug screening in patients ensuring patients receive the most effective treatment at the earliest time point.
Collapse
Affiliation(s)
- Xiaona Wen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University Nashville TN 37235 USA
| | - Yu-Chuan Ou
- Department of Chemical and Biomolecular Engineering, Vanderbilt University Nashville TN 37235 USA
| | - Galina Bogatcheva
- Department of Medicine, Vanderbilt University Medical Center Nashville TN 37232 USA
| | - Giju Thomas
- Vanderbilt Biophotonics Center, Vanderbilt University Nashville TN 37232 USA
| | | | - Bhuminder Singh
- Department of Medicine, Vanderbilt University Medical Center Nashville TN 37232 USA
| | - Eugene C Lin
- Department of Chemistry and Biochemistry, National Chung Cheng University Chiayi 62106 Taiwan
| | - Rizia Bardhan
- Department of Chemical and Biological Engineering, Iowa State University Ames IA 50012 USA
- Nanovaccine Institute, Iowa State University Ames IA 50012 USA
| |
Collapse
|
19
|
Lei S, Xu H, Chen N, Pan H, Xie W, He Y, Jin J. MKP-1 overexpression is associated with chemoresistance in bladder cancer via the MAPK pathway. Oncol Lett 2020; 20:1743-1751. [PMID: 32724417 PMCID: PMC7377201 DOI: 10.3892/ol.2020.11741] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 01/14/2020] [Indexed: 01/04/2023] Open
Abstract
Mitogen activated protein kinase phosphatase-1 (MKP-1) has been revealed to be overexpressed in bladder cancer, particularly in non-muscle invasive bladder cancer. MKP-1 may also be associated with chemotherapy resistance. However, the underlying mechanism is yet to be elucidated. The current study investigated the expression of MKP-1 by performing immunohistochemistry in surgically resected specimens obtained from primary and recurrent patients with bladder cancer. The results revealed that MKP-1 expression increased in recurrent patients. Additionally, a 3D model of the human bladder cancer cell line, RT112, was established to determine the role of MKP-1 in drug resistance. The results demonstrated that MKP-1 overexpression protected bladder cancer cells against cell death. Contrarily, MKP-1 knockdown was revealed to sensitize cells to death. In addition, the application of MAPK inhibitors effectively increased RT112 cell sensitivity to pirarubicin. In conclusion, the results of the current study indicated that MKP-1 treatment resulted in bladder cancer cell chemoresistance via JNK, ERK and p38 pathways. MKP-1 may also serve as a potential therapeutic target for chemoresistance in patients with bladder cancer.
Collapse
Affiliation(s)
- Siyu Lei
- Department of Urology, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China.,Department of Surgery, The 2nd Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, P.R. China
| | - Hong Xu
- Department of Urology, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China.,Department of Surgery, The 2nd Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, P.R. China
| | - Naiwen Chen
- Department of Urology, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China.,Department of Surgery, The 2nd Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, P.R. China
| | - Huan Pan
- Department of Central Laboratory, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Wenhua Xie
- Department of Urology, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Yi He
- Department of Urology, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Jing Jin
- Department of Urology, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| |
Collapse
|
20
|
Bao M, Zhang K, Wei Y, Hua W, Gao Y, Li X, Ye L. Therapeutic potentials and modulatory mechanisms of fatty acids in bone. Cell Prolif 2020; 53:e12735. [PMID: 31797479 PMCID: PMC7046483 DOI: 10.1111/cpr.12735] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/02/2019] [Accepted: 11/05/2019] [Indexed: 02/05/2023] Open
Abstract
Bone metabolism is a lifelong process that includes bone formation and resorption. Osteoblasts and osteoclasts are the predominant cell types associated with bone metabolism, which is facilitated by other cells such as bone marrow mesenchymal stem cells (BMMSCs), osteocytes and chondrocytes. As an important component in our daily diet, fatty acids are mainly categorized as long-chain fatty acids including polyunsaturated fatty acids (LCPUFAs), monounsaturated fatty acids (LCMUFAs), saturated fatty acids (LCSFAs), medium-/short-chain fatty acids (MCFAs/SCFAs) as well as their metabolites. Fatty acids are closely associated with bone metabolism and associated bone disorders. In this review, we summarized the important roles and potential therapeutic implications of fatty acids in multiple bone disorders, reviewed the diverse range of critical effects displayed by fatty acids on bone metabolism, and elucidated their modulatory roles and mechanisms on specific bone cell types. The evidence supporting close implications of fatty acids in bone metabolism and disorders suggests fatty acids as potential therapeutic and nutritional agents for the treatment and prevention of metabolic bone diseases.
Collapse
Affiliation(s)
- Minyue Bao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Kaiwen Zhang
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yangyini Wei
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Weihan Hua
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yanzi Gao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Xin Li
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Ling Ye
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesDepartment of Cariology and EndodonticsWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
21
|
Tavakol S, Ashrafizadeh M, Deng S, Azarian M, Abdoli A, Motavaf M, Poormoghadam D, Khanbabaei H, Afshar EG, Mandegary A, Pardakhty A, Yap CT, Mohammadinejad R, Kumar AP. Autophagy Modulators: Mechanistic Aspects and Drug Delivery Systems. Biomolecules 2019; 9:E530. [PMID: 31557936 PMCID: PMC6843293 DOI: 10.3390/biom9100530] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 09/14/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy modulation is considered to be a promising programmed cell death mechanism to prevent and cure a great number of disorders and diseases. The crucial step in designing an effective therapeutic approach is to understand the correct and accurate causes of diseases and to understand whether autophagy plays a cytoprotective or cytotoxic/cytostatic role in the progression and prevention of disease. This knowledge will help scientists find approaches to manipulate tumor and pathologic cells in order to enhance cellular sensitivity to therapeutics and treat them. Although some conventional therapeutics suffer from poor solubility, bioavailability and controlled release mechanisms, it appears that novel nanoplatforms overcome these obstacles and have led to the design of a theranostic-controlled drug release system with high solubility and active targeting and stimuli-responsive potentials. In this review, we discuss autophagy modulators-related signaling pathways and some of the drug delivery strategies that have been applied to the field of therapeutic application of autophagy modulators. Moreover, we describe how therapeutics will target various steps of the autophagic machinery. Furthermore, nano drug delivery platforms for autophagy targeting and co-delivery of autophagy modulators with chemotherapeutics/siRNA, are also discussed.
Collapse
Affiliation(s)
- Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Milad Ashrafizadeh
- Department of basic science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Maryam Azarian
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
- Departament de Bioquímica i Biologia Molecular, Institut de Biotecnologia i Biomedicina (IBB), Universitat Autónoma de Barcelona, Barcelona, Spain.
| | - Asghar Abdoli
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| | - Mahsa Motavaf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Delaram Poormoghadam
- Department of Medical Nanotechnology, Faculty of Advanced Sciences & Technology, Pharmaceutical Sciences Branch, Islamic Azad University, (IAUPS), Tehran, Iran.
| | - Hashem Khanbabaei
- Medical Physics Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Elham Ghasemipour Afshar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Ali Mandegary
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Abbas Pardakhty
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Celestial T Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.
| |
Collapse
|
22
|
Siraj AK, Begum R, Melosantos R, Albalawy W, Abboud J, Siraj N, Al-Kuraya KS. Zamzam water protects cancer cells from chemotherapy-induced apoptosis via mitogen-activated protein kinase-dependent pathway. Biomed Pharmacother 2019; 118:109376. [PMID: 31545262 DOI: 10.1016/j.biopha.2019.109376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/16/2019] [Accepted: 08/22/2019] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Many Muslims believe that water from the Zamzam well is the cure for every disease. Zamzam water (ZW) is naturally alkaline water consumed by millions of people worldwide. The current study investigated the effect of ZW on cell viability and apoptosis in breast, colorectal and ovarian cancer cell lines and compared the effect with that of drinking water (DW). METHODS Three different ZW samples collected from different sources at different periods were used. To balance the tonicity, ZW and DW were buffered using PBS and the pH was adjusted to 7.4. For the treatment, ZW and DW were diluted to 50% with RPMI medium (10% FBS). Cancer cell lines were treated with ZW or DW, with and without chemotherapeutic agents, for 24 h. Apoptosis was measured using flow cytometry whilst the level of protein expression was determined by Western blotting. RESULTS The results showed that treatment with ZW significantly increased cell proliferation compared to DW control. Treatment with ZW significantly suppressed the effect of chemotherapeutic agents on decreasing cell viability and inducing apoptosis in all the cancer cell lines compared to chemotherapeutic agents alone treated in DW. Furthermore, ZW treatment increased the phosphorylation of CRAF, MEK1/2, ERK1/2 and P38 proteins in these cell lines. Notably, treatment with ZW suppressed the effect of chemotherapy-induced reduction of CRAF, MEK1/2, ERK1/2 and P38 phosphorylation in breast and ovarian cancer cell lines. We also showed that silencing of ERK1/2 significantly increased the chemotherapy-induced apoptosis in breast and colorectal cancer cell lines. These data suggest that MAPK proteins; especially activated ERK1/2 may play a role in ZW mediated suppression of chemotherapy-induced cell death. CONCLUSIONS These findings clearly demonstrate that ZW protects cancer cells from chemotherapy-induced apoptosis through activation of the ERK1/2-MAPK signaling pathway.
Collapse
Affiliation(s)
- Abdul Khalid Siraj
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Rafia Begum
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Roxanne Melosantos
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Wafaa Albalawy
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Jehan Abboud
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Nabil Siraj
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Khawla S Al-Kuraya
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, P.O. Box 3354, Riyadh, 11211, Saudi Arabia.
| |
Collapse
|
23
|
Martino E, Vuoso DC, D'Angelo S, Mele L, D'Onofrio N, Porcelli M, Cacciapuoti G. Annurca apple polyphenol extract selectively kills MDA-MB-231 cells through ROS generation, sustained JNK activation and cell growth and survival inhibition. Sci Rep 2019; 9:13045. [PMID: 31506575 PMCID: PMC6736874 DOI: 10.1038/s41598-019-49631-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/23/2019] [Indexed: 01/15/2023] Open
Abstract
Polyphenols represent the most studied class of nutraceuticals that can be therapeutics for a large spectrum of diseases, including cancer. In this study, we investigated for the first time the antitumor activities of polyphenol extract from Annurca apple (APE) in MDA-MB-231 triple negative breast cancer cells, and we explored the underlying mechanisms. APE selectively inhibited MDA-MB-231 cell viability and caused G2/M phase arrest associated with p27 and phospho-cdc25C upregulation and with p21 downregulation. APE promoted reactive oxygen species (ROS) generation in MDA-MB-231 cells while it acted as antioxidant in non-tumorigenic MCF10A cells. We demonstrated that ROS generation represented the primary step of APE antitumor activity as pretreatment with antioxidant N-acetylcysteine (NAC) prevented APE-induced G2/M phase arrest, apoptosis, and autophagy. APE downregulated Dusp-1 and induced a significant increase in JNK/c-Jun phosphorylation that were both prevented by NAC. Moreover, downregulation of JNK by its specific inhibitor SP600125 significantly diminished the anticancer activity of APE indicating that ROS generation and sustained JNK activation represented the main underlying mechanism of APE-induced cell death. APE also inhibited AKT activation and downregulated several oncoproteins, such as NF-kB, c-myc, and β-catenin. In light of these results, APE may be an attractive candidate for drug development against triple negative breast cancer.
Collapse
Affiliation(s)
- Elisa Martino
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", via Luigi De Crecchio 7, 80138, Naples, Italy
| | - Daniela Cristina Vuoso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", via Luigi De Crecchio 7, 80138, Naples, Italy
| | - Stefania D'Angelo
- Department of Motor Sciences and Wellness, "Parthenope" University, via Medina 40, 80133, Naples, Italy
| | - Luigi Mele
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", via Luciano Armanni 5, 80138, Naples, Italy
| | - Nunzia D'Onofrio
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", via Luigi De Crecchio 7, 80138, Naples, Italy
| | - Marina Porcelli
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", via Luigi De Crecchio 7, 80138, Naples, Italy
| | - Giovanna Cacciapuoti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", via Luigi De Crecchio 7, 80138, Naples, Italy.
| |
Collapse
|
24
|
GP78 Cooperates with Dual-Specificity Phosphatase 1 To Stimulate Epidermal Growth Factor Receptor-Mediated Extracellular Signal-Regulated Kinase Signaling. Mol Cell Biol 2019; 39:MCB.00485-18. [PMID: 31061093 DOI: 10.1128/mcb.00485-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/20/2019] [Indexed: 12/15/2022] Open
Abstract
GP78 is an autocrine motility factor (AMF) receptor (AMFR) with E3 ubiquitin ligase activity that plays a significant role in tumor cell proliferation, motility, and metastasis. Aberrant extracellular signal-regulated kinase (ERK) activation via receptor tyrosine kinases promotes tumor proliferation and invasion. The activation of GP78 leads to ERK activation, but its underlying mechanism is not fully understood. Here, we show that GP78 is required for epidermal growth factor receptor (EGFR)-mediated ERK activation. On one hand, GP78 interacts with and promotes the ubiquitination and subsequent degradation of dual-specificity phosphatase 1 (DUSP1), an endogenous negative regulator of mitogen-activated protein kinases (MAPKs), resulting in ERK activation. On the other hand, GP78 maintains the activation status of EGFR, as evidenced by the fact that EGF fails to induce EGFR phosphorylation in GP78-deficient cells. By the regulation of both EGFR and ERK activation, GP78 promotes cell proliferation, motility, and invasion. Therefore, this study identifies a previously unknown signaling pathway by which GP78 stimulates ERK activation via DUSP1 degradation to mediate EGFR-dependent cancer cell proliferation and invasion.
Collapse
|
25
|
Han J, Park JC, Kang HM, Byeon E, Yoon DS, Lee MC, Sayed AEDH, Hwang UK, Lee JS. Adverse effects, expression of defense-related genes, and oxidative stress-induced MAPK pathway in the benzo[α]pyrene-exposed rotifer Brachionus rotundiformis. AQUATIC TOXICOLOGY 2019; 210:188-195. [PMID: 30870665 DOI: 10.1016/j.aquatox.2019.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 02/07/2023]
Abstract
To examine the adverse effects of the benzo[α]pyrene (B[α]P), the monogonont rotifer Brachionus rotundiformis was exposed to various concentration of B[α]P (0 [control], 1, 10, and 100 μg/L) and measured life cycle parameters (e.g., mortality, fecundity [cumulated number of offspring], and lifespan), reactive oxygen species (ROS), antioxidant enzymatic activity of glutathione S-transferase (GST). In addition, defense-related transcripts (e.g., glutathione S-transferases [GSTs], ATP binding cassette [ABCs] transporters) and Western blot analysis of mitogen-activated protein kinase (MAPK) signaling pathway were investigated in B[α]P-exposed rotifer. In this study, the total intracellular ROS level and GST activity were significantly increased (P < 0.05), while fecundity and lifespan were also significantly (P < 0.05) reduced in a concentration dependent manner in B[α]P-exposed B. rotundiformis. In addition, transcriptional regulation of GSTs and ABC transporters were significantly upregulated and downregulated (P < 0.05), respectively, suggesting that B[α]P can induce oxidative stress leading to induction of antioxidant system and detoxification mechanism. In addition to detoxification-related genes, B[α]P-exposed B. rotundiformis showed the increased levels of the p-JNK and p-p38, suggesting that B[α]P can activate MAPK signaling pathway in B. rotundiformis.
Collapse
Affiliation(s)
- Jeonghoon Han
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Jun Chul Park
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Hye-Min Kang
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Eunjin Byeon
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Deok-Seo Yoon
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Min-Chul Lee
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Alaa El-Din H Sayed
- Department of Zoology, Faculty of Science, Assiut University, 71516 Assiut, Egypt
| | - Un-Ki Hwang
- Marine Ecological Risk Assessment Center, West Sea Fisheries Research Institute, National Institute of Fisheries Science, Incheon 46083, South Korea
| | - Jae-Seong Lee
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
26
|
Fang Y, Yuan Y, Zhang LL, Lu JW, Feng JF, Hu SN. Downregulated GBX2 gene suppresses proliferation, invasion and angiogenesis of breast cancer cells through inhibiting the Wnt/β-catenin signaling pathway. Cancer Biomark 2019; 23:405-418. [PMID: 30223390 DOI: 10.3233/cbm-181466] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Gastrulation brain homeobox 2 (GBX2), a gene involved in mid/hindbrain region, has been revealed as one of the oncogene associated with certain cancers, as an example being prostate cancer. However, despite years of worldwide research, the underlying mechanism of GBX2 as well as its significance in breast cancer still remains unclear. Therefore, the present study evaluates the abilities of GBX gene silencing providing for the proliferation, invasion and angiogenesis of breast cancer cells by way of the Wnt/β-catenin signaling pathway. METHODS We employed a microarray analysis to screen out differentially expressed genes relative to breast cancer. Moreover, we retrieved GBX2 expression in breast cancer to find out the relationship between GBX2 expression and prognosis in breast cancer. We performed RT-qPCR to screen out cell lines with high GBX2 expression. Subsequently, both RT-qPCR and western blot analysis were employed so as to measure the combination of the mRNA and protein expressions of GBX2, β-catenin, vascular endothelial growth factor (VEGF), matrix metalloproteinase (MMP)-2, and MMP-9. The effect that GBX2 gene silencing and the Wnt/β-catenin signaling pathway had on cell proliferation, invasion, angiogenesis, and tumorigenic ability were evaluated. RESULTS GBX2 gene was also identified having played a role in breast cancer development due to its association with the Wnt/β-catenin signaling pathway. GBX2 gene silencing was found to be an inhibitor for the mRNA and protein expressions regulating β-catenin, VEGF, MMP-2, and MMP-9. Cell proliferation, invasion, angiogenesis, as well as tumorigenic ability in breast cancer were investigated and found to have been suppressed by the GBX2 gene silencing or inactivation of the Wnt/β-catenin signaling pathway. CONCLUSION The study has made an attempt to provide evidence to the idea that GBX2 gene silencing has an inhibition effect on the proliferation, invasion and angiogenesis of the breast cancer cells by inhibiting the activation of the Wnt/β-catenin signaling pathway.
Collapse
|
27
|
Buiga P, Elson A, Tabernero L, Schwartz JM. Modelling the role of dual specificity phosphatases in herceptin resistant breast cancer cell lines. Comput Biol Chem 2019; 80:138-146. [PMID: 30952040 DOI: 10.1016/j.compbiolchem.2019.03.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 03/23/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Breast cancer remains the most lethal type of cancer for women. A significant proportion of breast cancer cases are characterised by overexpression of the human epidermal growth factor receptor 2 protein (HER2). These cancers are commonly treated by Herceptin (Trastuzumab), but resistance to drug treatment frequently develops in tumour cells. Dual-specificity phosphatases (DUSPs) are thought to play a role in the mechanism of resistance, since some of them were reported to be overexpressed in tumours resistant to Herceptin. RESULTS We used a systems biology approach to investigate how DUSP overexpression could favour cell proliferation and to predict how this mechanism could be reversed by targeted inhibition of selected DUSPs. We measured the expression of 20 DUSP genes in two breast cancer cell lines following long-term (6 months) exposure to Herceptin, after confirming that these cells had become resistant to the drug. We constructed several Boolean models including specific substrates of each DUSP, and showed that our models correctly account for resistance when overexpressed DUSPs were kept activated. We then simulated inhibition of both individual and combinations of DUSPs, and determined conditions under which the resistance could be reversed. CONCLUSIONS These results show how a combination of experimental analysis and modelling help to understand cell survival mechanisms in breast cancer tumours, and crucially enable us to generate testable predictions potentially leading to new treatments of resistant tumours.
Collapse
Affiliation(s)
- Petronela Buiga
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel; School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Lydia Tabernero
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jean-Marc Schwartz
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
28
|
Cui Q, Xing J, Gu Y, Nan X, Ma W, Chen Y, Zhao H. GXYLT2 accelerates cell growth and migration by regulating the Notch pathway in human cancer cells. Exp Cell Res 2019; 376:1-10. [PMID: 30716301 DOI: 10.1016/j.yexcr.2019.01.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 01/27/2019] [Accepted: 01/31/2019] [Indexed: 12/14/2022]
Abstract
Glucoside xylosyltransferase2 (GXYLT2), a member of the human α-1,3-D-xylosyltransferases, functions to modify the first xylose to the O-Glucose residue on epidermal growth factor (EGF) repeats of Notch receptors. It is well-established that the Notch signaling pathway plays a critical role in proper development and homeostasis. However, the regulatory role of EGF xylosylation in Notch signaling and different cell activities in human cells remains unknown. In this study, we showed that knockdown of GXYLT2 suppressed human cell proliferation and induced G1/S phase cell cycle arrest. GXYLT2 downregulation also inhibited cell migration and invasion, whereas the overexpression of GXYLT2 had the opposite effects. Additionally, GXYLT2 activated Notch signaling and promoted the phosphorylation of MAPKs but not PI3K and Akt. Taken together, our findings indicated that GXYLT2 plays an important role in cell activities via regulation of the Notch signaling.
Collapse
Affiliation(s)
- Qi Cui
- Department of Medical Genetics, Peking University School of Basic Medical Sciences, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Jinhao Xing
- Department of Medical Genetics, Peking University School of Basic Medical Sciences, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Yajuan Gu
- Department of Medical Genetics, Peking University School of Basic Medical Sciences, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Xu Nan
- Department of Medical Genetics, Peking University School of Basic Medical Sciences, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Wenping Ma
- Department of Medical Genetics, Peking University School of Basic Medical Sciences, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Yingyu Chen
- Peking University Center for Human Disease Genomics, Beijing 100191, China; Department of Immunology, Peking University School of Basic Medical Science, Key Laboratory of Medical Immunology, Beijing 100191, China
| | - Hongshan Zhao
- Department of Medical Genetics, Peking University School of Basic Medical Sciences, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China.
| |
Collapse
|
29
|
Hypoxia-inducible factors promote breast cancer stem cell specification and maintenance in response to hypoxia or cytotoxic chemotherapy. Adv Cancer Res 2019; 141:175-212. [PMID: 30691683 DOI: 10.1016/bs.acr.2018.11.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Clinical studies have revealed that breast cancers contain regions of intratumoral hypoxia (reduced oxygen availability), which activates hypoxia-inducible factors (HIFs). The relationship between intratumoral hypoxia, distant metastasis and cancer mortality has been well established. A major mechanism by which intratumoral hypoxia contributes to disease progression is through induction of the breast cancer stem cell (BCSC) phenotype. BCSCs are a small subpopulation of cells with the capability for self-renewal. BCSCs have been implicated in resistance to chemotherapy, disease recurrence, and metastasis. In this review, we will discuss our current understanding of the molecular mechanisms underlying HIF-dependent induction of the BCSC phenotype in response to hypoxia or chemotherapy.
Collapse
|
30
|
The Modulatory Role of MicroRNA-873 in the Progression of KRAS-Driven Cancers. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 14:301-317. [PMID: 30654191 PMCID: PMC6348737 DOI: 10.1016/j.omtn.2018.11.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 12/15/2022]
Abstract
KRAS is one of the most frequently mutated proto-oncogenes in pancreatic ductal adenocarcinoma (PDAC) and aberrantly activated in triple-negative breast cancer (TNBC). A profound role of microRNAs (miRNAs) in the pathogenesis of human cancer is being uncovered, including in cancer therapy. Using in silico prediction algorithms, we identified miR-873 as a potential regulator of KRAS, and we investigated its role in PDAC and TNBC. We found that reduced miR-873 expression is associated with shorter patient survival in both cancers. miR-873 expression is significantly repressed in PDAC and TNBC cell lines and inversely correlated with KRAS levels. We demonstrate that miR-873 directly bound to the 3′ UTR of KRAS mRNA and suppressed its expression. Notably, restoring miR-873 expression induced apoptosis; recapitulated the effects of KRAS inhibition on cell proliferation, colony formation, and invasion; and suppressed the activity of ERK and PI3K/AKT, while overexpression of KRAS rescued the effects mediated by miR-873. Moreover, in vivo delivery of miR-873 nanoparticles inhibited KRAS expression and tumor growth in PDAC and TNBC tumor models. In conclusion, we provide the first evidence that miR-873 acts as a tumor suppressor by targeting KRAS and that miR-873-based gene therapy may be a therapeutic strategy in PDAC and TNBC.
Collapse
|
31
|
Teng F, Xu Z, Chen J, Zheng G, Zheng G, Lv H, Wang Y, Wang L, Cheng X. DUSP1 induces apatinib resistance by activating the MAPK pathway in gastric cancer. Oncol Rep 2018; 40:1203-1222. [PMID: 29956792 PMCID: PMC6072387 DOI: 10.3892/or.2018.6520] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 06/13/2018] [Indexed: 12/13/2022] Open
Abstract
Dual-specificity phosphatase-1 (DUSP1) is an oncogene that is associated with cancer progression following drug resistance. In order to investigate the potential relationship between DUSP1 and apatinib resistance in gastric cancer cells, we preformed many assays to study this problem. DUSP1 gene was detected by RT-qPCR assay, proteins in MAPK pathway were quantified by western blot assay, and CCK-8 assay, flow cytometry and Hoechest 33342 stain were performed to detect the resistance of cells, cell cycles and apoptosis, respectively. Immunohistochemical staining was used to discover the expression of DUSP1 protein in patients' tumor or paratumor tissues. It was found that apatinib (Apa)-resistant gastric cancer (GC) cells showed increased expression of DUSP1, whereas the knockdown of DUSP1 in resistant cells resensitized these cells to Apa. The restored sensitivity to Apa was the result of inactivation of mitogen-activated protein kinase (MAPK) signaling and the induction of apoptosis. The in vitro use of Apa in combination with a DUSP1 inhibitor, triptolide, exerted significant effects on inhibiting the expression of DUSP1, growth inhibition, and apoptosis via the inactivation of MAPK signaling. In patients who did not undergo chemotherapy or targeted therapy, the expression of DUSP1 in adjacent tissues was higher when compared with that observed in tumor tissues. In addition, the expression of DUSP1 was higher in the early stages of GC than in the advanced stages. The expression of DUSP1 in tumor tissues was not associated with the survival rate of the patients. Therefore, increased expression of DUSP1 may be responsible for Apa resistance, and DUSP1 may serve as a biomarker for Apa efficacy. In conclusion, inducing the downregulation of DUSP1 may be a promising strategy to overcome Apa resistance.
Collapse
Affiliation(s)
- Fei Teng
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Zhiyuan Xu
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diagnosis and Treatment of Digestive System Tumor, Hangzhou, Zhejiang 310006, P.R. China
| | - Jiahui Chen
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Guowei Zheng
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Guodian Zheng
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diagnosis and Treatment of Digestive System Tumor, Hangzhou, Zhejiang 310006, P.R. China
| | - Hang Lv
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diagnosis and Treatment of Digestive System Tumor, Hangzhou, Zhejiang 310006, P.R. China
| | - Yiping Wang
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diagnosis and Treatment of Digestive System Tumor, Hangzhou, Zhejiang 310006, P.R. China
| | - Lijing Wang
- Department of Medical Imaging, Zhejiang Provincial Tumor Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Xiangdong Cheng
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diagnosis and Treatment of Digestive System Tumor, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
32
|
Lu H, Tran L, Park Y, Chen I, Lan J, Xie Y, Semenza GL. Reciprocal Regulation of DUSP9 and DUSP16 Expression by HIF1 Controls ERK and p38 MAP Kinase Activity and Mediates Chemotherapy-Induced Breast Cancer Stem Cell Enrichment. Cancer Res 2018; 78:4191-4202. [PMID: 29880481 DOI: 10.1158/0008-5472.can-18-0270] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/08/2018] [Accepted: 06/01/2018] [Indexed: 02/05/2023]
Abstract
Triple-negative breast cancer (TNBC) has a poor prognosis due to its aggressive characteristics and lack of targeted therapies. Cytotoxic chemotherapy may reduce tumor bulk, but leaves residual disease due to the persistence of chemotherapy-resistant breast cancer stem cells (BCSC), which are critical for tumor recurrence and metastasis. Here, we demonstrate that hypoxia-inducible factor (HIF)-1-dependent regulation of mitogen-activated protein kinase (MAPK) signaling pathways contributes to chemotherapy-induced BCSC enrichment. Chemotherapy increased DUSP9 expression and decreased DUSP16 expression in a HIF1-dependent manner, leading to inhibition of ERK and activation of p38 signaling pathways, respectively. Inhibition of ERK caused transcriptional induction of the pluripotency factor Nanog through decreased inactivating phosphorylation of FoxO3, while activation of p38 stabilized Nanog and Klf4 mRNA through increased inactivating phosphorylation of RNA-binding protein ZFP36L1, both of which promoted specification of the BCSC phenotype. Inhibition of HIF1 or p38 signaling blocked chemotherapy-induced pluripotency factor expression and BCSC enrichment. These surprising results delineate a mechanism by which a transcription factor switches cells from ERK to p38 signaling in response to chemotherapy and suggest that therapeutic targeting of HIF1 or the p38 pathway in combination with chemotherapy will block BCSC enrichment and improve outcome in TNBC.Significance: These findings provide a molecular mechanism that may account for the increased relapse rate of women with TNBC who are treated with cytotoxic chemotherapy and suggest that combining chemotherapy with an inhibitor of HIF1 or p38 activity may increase patient survival. Cancer Res; 78(15); 4191-202. ©2018 AACR.
Collapse
Affiliation(s)
- Haiquan Lu
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Linh Tran
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Youngrok Park
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ivan Chen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jie Lan
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yangyiran Xie
- Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Gregg L Semenza
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland. .,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
33
|
Qian CJ, Qi YX, Zhong S, Zeng JP, Chen XY, Yao J. Mitogen-activated protein kinase inhibition enhances the antitumor effects of sporamin in human pancreatic cancer cells. Oncol Lett 2018; 16:1237-1242. [PMID: 30061945 DOI: 10.3892/ol.2018.8746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/17/2018] [Indexed: 01/07/2023] Open
Abstract
Sporamin, a sweet potato tuber storage protein, is a Kunitz-type trypsin inhibitor (TI) that has exhibited antitumor activity through poorly defined mechanisms in a number of types of tumor cells. The present study aimed to analyze the combined effects of sporamin and three mitogen-activated protein kinase (MAPK) inhibitors, PD98059, SP600125 and SB203580, on the pancreatic cancer cell line, PANC-1. Cell proliferation activity was assessed using a 3H-thymidine incorporation assay, and cell viability was analyzed using an MTT assay. Apoptosis was assayed by flow cytometry and fluorescence microscopy. Protein expression levels in PANC-1 cells were determined by western blotting. The results of this analysis demonstrated that sporamin induced a temporary increase in the phosphorylation of MAPKs, including phosphorylated extracellular signal regulated-kinase 1/2, phosphorylated c-Jun amino-terminal protein kinase 1/2 and phosphorylated p38-MAPK, in a concentration-dependent manner. However, treatment with MAPK inhibitors promoted the inhibition of cell proliferation and viability, and the induction of apoptosis in sporamin-treated PANC-1 cells. In conclusion, the present study demonstrated that MAPK inhibition enhanced the antitumor activity of sporamin in PANC-1 cells.
Collapse
Affiliation(s)
- Cui-Juan Qian
- Department of Gastroenterology, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, Zhejiang 318000, P.R. China.,Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Yong-Xiao Qi
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Sheng Zhong
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Ju-Ping Zeng
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Xiao-Ying Chen
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Jun Yao
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| |
Collapse
|
34
|
Buiga P, Elson A, Tabernero L, Schwartz JM. Regulation of dual specificity phosphatases in breast cancer during initial treatment with Herceptin: a Boolean model analysis. BMC SYSTEMS BIOLOGY 2018; 12:11. [PMID: 29671404 PMCID: PMC5907139 DOI: 10.1186/s12918-018-0534-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Background 25% of breast cancer patients suffer from aggressive HER2-positive tumours that are characterised by overexpression of the HER2 protein or by its increased tyrosine kinase activity. Herceptin is a major drug used to treat HER2 positive breast cancer. Understanding the molecular events that occur when breast cancer cells are exposed to Herceptin is therefore of significant importance. Dual specificity phosphatases (DUSPs) are central regulators of cell signalling that function downstream of HER2, but their role in the cellular response to Herceptin is mostly unknown. This study aims to model the initial effects of Herceptin exposure on DUSPs in HER2-positive breast cancer cells using Boolean modelling. Results We experimentally measured expression time courses of 21 different DUSPs between 0 and 24 h following Herceptin treatment of human MDA-MB-453 HER2-positive breast cancer cells. We clustered these time courses into patterns of similar dynamics over time. In parallel, we built a series of Boolean models representing the known regulatory mechanisms of DUSPs and then demonstrated that the dynamics predicted by the models is in agreement with the experimental data. Furthermore, we used the models to predict regulatory mechanisms of DUSPs, where these mechanisms were partially known. Conclusions Boolean modelling is a powerful technique to investigate and understand signalling pathways. We obtained an understanding of different regulatory pathways in breast cancer and new insights on how these signalling pathways are activated. This method can be generalized to other drugs and longer time courses to better understand how resistance to drugs develops in cancer cells over time. Electronic supplementary material The online version of this article (10.1186/s12918-018-0534-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Petronela Buiga
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel.,School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Lydia Tabernero
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jean-Marc Schwartz
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
35
|
Wu Q, Finley SD. Predictive model identifies strategies to enhance TSP1-mediated apoptosis signaling. Cell Commun Signal 2017; 15:53. [PMID: 29258506 PMCID: PMC5735807 DOI: 10.1186/s12964-017-0207-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 12/07/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Thrombospondin-1 (TSP1) is a matricellular protein that functions to inhibit angiogenesis. An important pathway that contributes to this inhibitory effect is triggered by TSP1 binding to the CD36 receptor, inducing endothelial cell apoptosis. However, therapies that mimic this function have not demonstrated clear clinical efficacy. This study explores strategies to enhance TSP1-induced apoptosis in endothelial cells. In particular, we focus on establishing a computational model to describe the signaling pathway, and using this model to investigate the effects of several approaches to perturb the TSP1-CD36 signaling network. METHODS We constructed a molecularly-detailed mathematical model of TSP1-mediated intracellular signaling via the CD36 receptor based on literature evidence. We employed systems biology tools to train and validate the model and further expanded the model by accounting for the heterogeneity within the cell population. The initial concentrations of signaling species or kinetic rates were altered to simulate the effects of perturbations to the signaling network. RESULTS Model simulations predict the population-based response to strategies to enhance TSP1-mediated apoptosis, such as downregulating the apoptosis inhibitor XIAP and inhibiting phosphatase activity. The model also postulates a new mechanism of low dosage doxorubicin treatment in combination with TSP1 stimulation. Using computational analysis, we predict which cells will undergo apoptosis, based on the initial intracellular concentrations of particular signaling species. CONCLUSIONS This new mathematical model recapitulates the intracellular dynamics of the TSP1-induced apoptosis signaling pathway. Overall, the modeling framework predicts molecular strategies that increase TSP1-mediated apoptosis, which is useful in many disease settings.
Collapse
Affiliation(s)
- Qianhui Wu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Stacey D Finley
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA.
- Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
36
|
Sundaraneedi MK, Ammit AJ, Tedla BA, Pearson MS, Loukas A, Keene FR, Collins JG. Tetranuclear Polypyridylruthenium(II) Complexes as Inhibitors and Down-Regulators of Phosphatase Enzymes. ChemistrySelect 2017. [DOI: 10.1002/slct.201702118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Madhu K. Sundaraneedi
- School of Physical; Environmental & Mathematical Sciences; University of New South Wales; Australian Defence Force Academy; Canberra, ACT 2600 Australia
| | - Alaina J. Ammit
- Woolcock Emphysema Centre; Woolcock Institute of Medical Research; University of Sydney; Sydney, NSW 2006 Australia
- School of Life Sciences; University of Technology Sydney; Sydney, NSW 2007 Australia
| | - Bemnet A. Tedla
- Centre for Biodiscovery & Molecular Development of Therapeutics/Australian Institute for Tropical Health & Medicine; James Cook University; Cairns, QLD 4878 Australia
| | - Mark S. Pearson
- Centre for Biodiscovery & Molecular Development of Therapeutics/Australian Institute for Tropical Health & Medicine; James Cook University; Cairns, QLD 4878 Australia
| | - Alex Loukas
- Centre for Biodiscovery & Molecular Development of Therapeutics/Australian Institute for Tropical Health & Medicine; James Cook University; Cairns, QLD 4878 Australia
| | - F. Richard Keene
- Centre for Biodiscovery & Molecular Development of Therapeutics/Australian Institute for Tropical Health & Medicine; James Cook University; Cairns, QLD 4878 Australia
- School of Physical Sciences; University of Adelaide; Adelaide, SA 5005 Australia
| | - J. Grant Collins
- School of Physical; Environmental & Mathematical Sciences; University of New South Wales; Australian Defence Force Academy; Canberra, ACT 2600 Australia
| |
Collapse
|
37
|
Kuo SH, Yang SY, You SL, Lien HC, Lin CH, Lin PH, Huang CS. Polymorphisms of ESR1, UGT1A1, HCN1, MAP3K1 and CYP2B6 are associated with the prognosis of hormone receptor-positive early breast cancer. Oncotarget 2017; 8:20925-20938. [PMID: 28178648 PMCID: PMC5400556 DOI: 10.18632/oncotarget.14995] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 01/03/2017] [Indexed: 12/22/2022] Open
Abstract
In this study, we investigated whether single nucleotide polymorphisms (SNPs) identified by genome-wide association study (GWAS) (MAP3K1, FGFR2, TNRC9, HCN1, and 5p12), and SNPs involved in the metabolism of estrogen (CYP19, COMT, ESR1, and UGT1A1), tamoxifen (CYP2C9, CYP2C19, CYP3A5, and CYP2D6), and chemotherapeutic agents (ABCB1, ALDH3A1, and CYP2B6) are associated with the prognoses of 414 hormone receptor (HR)-positive early breast cancers with negative or 1 to 3 nodal metastases. At a median follow-up period of 10.6 years, 363 patients were alive, and 51 (12.3%) had died. Multiple-adjusted hazard ratios (aHRs) and the corresponding 95% confidence intervals for distant disease-free survival (DDFS), disease-free survival (DFS), and overall survival (OS) in association with the genotypes of 34 SNPs from the above-mentioned 16 genes were evaluated, using the stepwise selection Cox model. We found that the SNP, ESR1-codon325 rs1801132 (G/G+G/C), was associated with a longer DDFS, whereas UGT1A1 rs4148323 (A/A+A/G), and HCN1 rs981782 (A/A+A/C) were significantly associated with poorer DDFS. MAP3K1 rs889312 (C/C) and CYP2B6 rs3211371 (T/C) were significantly associated with poor DFS, DDFS and OS. Among premenopausal women, MAP3K1 rs889312 (C/C), CYP2B6 rs3211371 (T/C), CYP2B6 rs4802101 (T/T), ABCB1 rs2032582 (C/C), and ALDH3A1 rs2231142 (G/G) were significantly associated with poor DDFS, DFS, or OS. Our results provide additional evidence that genetic polymorphisms observed in SNPs are associated with the prognoses of patients with HR-positive breast cancers; this may indicate different treatment strategies for these patients.
Collapse
Affiliation(s)
- Sung-Hsin Kuo
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.,Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.,Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan.,National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shi-Yi Yang
- Graduate Institute of Epidemiology, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - San-Lin You
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan.,Big Data Research Center, Fu-Jen Catholic University, New Taipei, Taiwan
| | - Huang-Chun Lien
- Department of Pathology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Hung Lin
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.,Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Po-Han Lin
- Department of Medical Genetics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chiun-Sheng Huang
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
38
|
The Role of the MAPK Signaling, Topoisomerase and Dietary Bioactives in Controlling Cancer Incidence. Diseases 2017; 5:diseases5020013. [PMID: 28933366 PMCID: PMC5547980 DOI: 10.3390/diseases5020013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/16/2017] [Accepted: 04/19/2017] [Indexed: 12/16/2022] Open
Abstract
Reactive oxygen species (ROS) are common products of mitochondrial oxidative phosphorylation, xenobiotics metabolism and are generated in response to several environmental stress conditions. Some of them play important biochemical roles in cellular signal transduction and gene transcription. On the other hand, ROS are known to be involved in a wide range of human diseases, including cancer. The excessive production of such ROS together with disruption of homeostasis detoxifying mechanisms can mediate a series of cellular oxidative stresses. The oxidative stress of redundant free radicals production can lead to oxidative denaturation of cellular macromolecules including proteins, lipids and DNA. Moreover, oxidative damage is one of the major causes of DNA mutations, replication errors and genomic abnormalities which result in either inhibition or induction of transcription, and end with the disturbance of signal transduction pathways. Among affected signaling pathways are redox-sensitive kinases. The stimulation of these kinases induces several transcription factors through the phosphorylation of their module proteins. The activation of such pathways induces proliferation and cellular transformation. A diet rich in antioxidant compounds has potential health benefits, and there is a growing interest in the role of natural antioxidants in nutrition for prevention and cure of cancer diseases. A controversy has risen regarding the relation between antioxidants and the significant decrease in the risk of cancer incidence. In this review, we will focus on redox-sensitive kinases signaling pathways, highlighting the effects of dietary antioxidant on the prevention, incidence, prognosis or even treatment of human cancers. In addition, we will place emphasis on the chemical classes of pterocarpans as natural anti-oxidants/cancers as well as their underlying mechanisms of action, including their effects on MAPKs and topoisomerase activities.
Collapse
|
39
|
Castrellon AB. Novel Strategies to Improve the Endocrine Therapy of Breast Cancer. Oncol Rev 2017; 11:323. [PMID: 28584571 PMCID: PMC5444409 DOI: 10.4081/oncol.2017.323] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 05/04/2017] [Indexed: 12/26/2022] Open
Abstract
Endocrine therapy (ET) constitutes the usual first-line of therapy for patients in the treatment of metastatic hormone receptor-positive breast cancer. Unfortunately, not all patients respond to first-line endocrine treatment due to intrinsic resistance, while others may initially respond but eventually progress with secondary acquired resistance leading to disease progression. Mechanisms of resistance to anti-estrogen therapy include, loss of expression for estrogen or progesterone receptor, upregulation of epidermal receptor growth factor 2, increased receptor tyrosine kinase signaling, leading to activation of various intracellular pathways that are involved in signal transduction such as PI3K/AKT/mammalian target of rapamycin, and others. Growing understanding of the signal cascade of estrogen receptors and the signaling pathways that interact with estrogen receptors has revealed the complex role of these receptors in cell growth and proliferation, and on the mechanism in development of resistance. These insights have led to the development of targeted therapies that may prove to be effective options for the treatment of breast cancer and may overcome hormone therapy resistance. In this review we summarize some of the mechanisms of endocrine resistance, selected clinical trials of ET and targeted therapies, which might interfere with estrogen receptor pathways and might reduce or reverse resistance to traditional, sequential, single-agent ET.
Collapse
Affiliation(s)
- Aurelio Bartolome Castrellon
- Medical Oncology, Breast Cancer Center, Memorial Cancer Institute, Memorial Healthcare System, Hollywood, FL, USA
| |
Collapse
|
40
|
Su X, Zhang L, Li H, Cheng P, Zhu Y, Liu Z, Zhao Y, Xu H, Li D, Gao H, Zhang T. MicroRNA-134 targets KRAS to suppress breast cancer cell proliferation, migration and invasion. Oncol Lett 2017; 13:1932-1938. [PMID: 28454346 DOI: 10.3892/ol.2017.5644] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 09/12/2016] [Indexed: 12/19/2022] Open
Abstract
The expression patterns and functions of microRNA-134 (miR-134) have been previously studied in numerous types of cancer. To the best of our knowledge, this is the first study of miR-134 in human breast cancer. In the present study, the expression patterns, biological functions and underlying molecular mechanisms of miR-134 in human breast cancer were investigated. Reverse transcription-quantitative polymerase chain reaction evaluated the expression of miR-134 in human breast cancer tissues, matched normal adjacent tissues, breast cancer cell lines and a normal mammary epithelial cell line. Following transfection with miR-134, an MTT assay, cell migration assay, cell invasion assay, western blot analysis and a luciferase assay were performed on the MCF-7 and MDA-MB-231 human breast cancer cell lines. The findings revealed that miR-134 expression levels were significantly downregulated in breast cancer cells. Statistical analysis demonstrated that low expression of miR-134 was significantly associated with lymph node metastasis, TNM stage and reduced cell differentiation. It was observed that miR-134 inhibited the growth, migration and invasion of breast cancer cells. Additionally, the present study indicated that miR-134 may directly target the Kirsten rat sarcoma viral oncogene homolog in breast cancer tissues. These results suggest that miR-134 may be used as a potential therapeutic biomarker in breast cancers.
Collapse
Affiliation(s)
- Xiaomei Su
- Department of Oncology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Ling Zhang
- Department of Oncology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Hua Li
- Department of Oncology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Peng Cheng
- Department of Oncology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Yajie Zhu
- Department of Oncology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Zhen Liu
- Department of Oncology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Yu Zhao
- Department of Oncology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Hongyu Xu
- Department of Oncology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Dong Li
- Department of Oncology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Hui Gao
- Department of Oncology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Tao Zhang
- Department of Oncology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| |
Collapse
|
41
|
Akinrinde AS, Omobowale O, Oyagbemi A, Asenuga E, Ajibade T. Protective effects of kolaviron and gallic acid against cobalt-chloride-induced cardiorenal dysfunction via suppression of oxidative stress and activation of the ERK signaling pathway. Can J Physiol Pharmacol 2016; 94:1276-1284. [DOI: 10.1139/cjpp-2016-0197] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cobalt (Co) toxicity is a potential public health problem due to recent renewed use of Co in orthopedic implants, dietary supplements, and blood doping in athletes and horses. We investigated the protective roles of kolaviron (KV), a bi-flavonoid of Garcinia kola, and gallic acid (GA) on cobalt chloride (CoCl2)-induced cardiorenal damage in rats. CoCl2 caused significant increases (p < 0.05) in serum creatine kinase–myocardial band (CK-MB), lactate dehydrogenase (LDH), aspartate transaminase (AST), xanthine oxidase (XO), urea, creatinine, malondialdehyde, H2O2, nitric oxide, as well as C-reactive protein expression, along with significant (p < 0.05) reduction in cardiac and renal expression of extracellular signal regulated kinase (ERK) and the activities of superoxide dismutase, catalase, and glutathione S-transferase. KV and GA prevented the toxic effects of CoCl2 by stimulating ERK expression and reversing Co-induced biochemical changes. Administration of CoCl2 alone did not significantly alter ECG patterns in the rats, although co-treatment with KV (200 mg/kg) produced QT-segment prolongation and also appeared to potentiate Co hypotension. Histopathology of the heart and kidneys of rats treated with KV and GA confirmed the biochemical data. KV and GA thus protected against cardiac and renal damage in Co intoxication via antioxidant and (or) cell survival mechanisms, possibly involving ERK activation.
Collapse
Affiliation(s)
- Akinleye Stephen Akinrinde
- Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, 900001 Nigeria
| | - Olutayo Omobowale
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, 900001 Nigeria
| | - Ademola Oyagbemi
- Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, 900001 Nigeria
| | - Ebunoluwa Asenuga
- Department of Veterinary Biochemistry, Faculty of Veterinary Medicine, University of Benin, Nigeria
| | - Temitayo Ajibade
- Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, 900001 Nigeria
| |
Collapse
|
42
|
Rincón R, Zazo S, Chamizo C, Manso R, González-Alonso P, Martín-Aparicio E, Cristóbal I, Cañadas C, Perona R, Lluch A, Eroles P, García-Foncillas J, Albanell J, Rovira A, Madoz-Gúrpide J, Rojo F. c-Jun N-Terminal Kinase Inactivation by Mitogen-Activated Protein Kinase Phosphatase 1 Determines Resistance to Taxanes and Anthracyclines in Breast Cancer. Mol Cancer Ther 2016; 15:2780-2790. [PMID: 27599524 DOI: 10.1158/1535-7163.mct-15-0920] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 08/07/2016] [Indexed: 11/16/2022]
Abstract
MAPK phosphatase-1 (MKP-1) is overexpressed during malignant transformation of the breast in many patients, and it is usually associated with chemoresistance through interference with JNK-driven apoptotic pathways. Although the molecular settings of the mechanism have been documented, details about the contribution of MKP-1 to the failure of chemotherapeutic interventions are unclear. Transient overexpression of MKP-1 and treatment with JNK-modulating agents in breast carcinoma cells confirmed the mediation of MKP-1 in the resistance to taxanes and anthracyclines in breast cancer, through the inactivation of JNK1/2. We next assessed MKP-1 expression and JNK1/2 phosphorylation status in a large cohort of samples from 350 early breast cancer patients treated with adjuvant anthracycline-based chemotherapy. We detected that MKP-1 overexpression is a recurrent event predominantly linked to dephosphorylation of JNK1/2 with an adverse impact on relapse of the tumor and overall and disease-free survival. Moreover, MKP-1 and p-JNK1/2 determinations in 64 locally advanced breast cancer patients treated with neoadjuvant taxane-based chemotherapy showed an inverse correlation between MKP-1 overexpression (together with JNK1/2 inhibition) and the pathologic response of the tumors. Our results emphasize the importance of MKP-1 as a potential predictive biomarker for a subset of breast cancer patients with worse outcome and less susceptibility to treatment. Mol Cancer Ther; 15(11); 2780-90. ©2016 AACR.
Collapse
Affiliation(s)
- Raúl Rincón
- Pathology Department, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - Sandra Zazo
- Pathology Department, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - Cristina Chamizo
- Pathology Department, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - Rebeca Manso
- Pathology Department, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | | | | | - Ion Cristóbal
- Translational Oncology Division, Oncohealth Institute, Health Research Institute FJD-UAM, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Carmen Cañadas
- Pathology Department, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - Rosario Perona
- "Alberto Sols" Biomedical Research Institute CSIC-UAM, Madrid, Spain
| | - Ana Lluch
- Institute of Health Research INCLIVA, Valencia, Spain
| | - Pilar Eroles
- Institute of Health Research INCLIVA, Valencia, Spain
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, Health Research Institute FJD-UAM, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Joan Albanell
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Ana Rovira
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona, Spain
| | | | - Federico Rojo
- Pathology Department, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain.
| |
Collapse
|
43
|
Yeh DYW, Wu CC, Chin YP, Lu CJ, Wang YH, Chen MC. Mechanisms of human lymphotoxin beta receptor activation on upregulation of CCL5/RANTES production. Int Immunopharmacol 2015; 28:220-9. [DOI: 10.1016/j.intimp.2015.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 06/05/2015] [Accepted: 06/05/2015] [Indexed: 11/28/2022]
|
44
|
Choi EK, Yeo JS, Park CY, Na HI, Lim JA, Lee JE, Hong SW, Park SS, Lim DG, Kwak KH. Inhibition of reactive oxygen species downregulates the MAPK pathway in rat spinal cord after limb ischemia reperfusion injury. Int J Surg 2015; 22:74-8. [PMID: 26283297 DOI: 10.1016/j.ijsu.2015.08.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/06/2015] [Accepted: 08/05/2015] [Indexed: 01/19/2023]
Abstract
INTRODUCTION We examined the activity of mitogen-activated protein kinase (MAPK) family members, extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38, in rats pinal cord after hind limb ischemia reperfusion (IR) and analyzed the role of reactive oxygen species (ROS) as mediators of MAPK signaling under these conditions. METHODS In experiment 1, hind limb IR rats were treated intraperitoneally with one of following agents at 30 min before reperfusion: allopurinol (4, 40 mg/kg), superoxide dismutase (SOD, 4000 U/kg), N-nitro-l-arginine methyl ester (l-NAME, 10 mg/kg), or SOD (4000 U/kg) + l-NAME (10 mg/kg). In experiment 2, 5,10,15,20-tetrakis (N-methyl-4'-pyridyl) porphyrinato iron (III) (FeTMPyP) was administered intraperitoneally (1, 3, or 10 mg/kg) 30 min before reperfusion. After 3 d reperfusion period, the spinal cord (L4-6) was harvested to investigate MAPK signaling activity. RESULTS In experiment 1, p-ERK and p-JNK levels were significantly higher in the IR group than sham group. Administration of allopurinol, SOD, l-NAME, or SOD + l-NAME significantly reduced the IR-induced increase in p-ERK and p-JNK levels. There were no significant differences in p-p38 levels. In experiment 2, FeTMPyP significantly reduced the IR-induced increase in p-ERK and p-JNK levels in a dose-dependent manner. CONCLUSIONS Activation of ERK and JNK in the spinal cord was induced by hind limb IR and was not accompanied by p38 activation. IR-induced MAPK phosphorylation was reduced by inhibition of superoxide, nitric oxide, and peroxynitrite, indicating that ROS produced by hind limb IR mediate the activation of these signaling pathways in the spinal cord, potentially affecting distant organs.
Collapse
Affiliation(s)
- Eun Kyung Choi
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Jin-Seok Yeo
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Chan Yoon Park
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Ho in Na
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Jung a Lim
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Jeong-Eun Lee
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Seong Wook Hong
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Sung-Sik Park
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Dong Gun Lim
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Kyung Hwa Kwak
- Department of Anesthesiology and Pain Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea.
| |
Collapse
|
45
|
Patel BS, Co WS, Donat C, Wang M, Che W, Prabhala P, Schuster F, Schulz V, Martin JL, Ammit AJ. Repression of breast cancer cell growth by proteasome inhibitors in vitro: impact of mitogen-activated protein kinase phosphatase 1. Cancer Biol Ther 2015; 16:780-9. [PMID: 25774547 DOI: 10.1080/15384047.2015.1026465] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mitogen activated protein kinase phosphatase-1 (MKP-1) has emerged as an important protein mediating breast cancer oncogenesis and chemoresistance to cancer chemotherapies, especially proteasome inhibitors. In this in vitro study, we utilized the breast cancer epithelial cell lines MCF-7 and MDA-MB-231, in comparison to MCF-10A control cells, to examine the impact of MKP-1 on breast cancer cell growth and repression by proteasome inhibitors. We confirm that proteasome inhibitors MG-132 and bortezomib induce MKP-1 protein upregulation and we show that one of the ways in which bortezomib increases MKP-1 in breast cancer cells, in addition to inhibition of ubiquitin-proteasome system, is via upregulation of MKP-1 mRNA expression in p38 MAPK-mediated manner. Notably, these effects are specific to cancer cells, as bortezomib activated p38 MAPK and induced MKP-1 in MCF-7 and MDA-MB-231 breast cancer cells, but not in control cells (MCF-10A). We took a dual approach toward targeting MKP-1 to show that bortezomib-induced effects are enhanced. Firstly, treatment with the non-specific MKP-1 inhibitor triptolide reduces breast cancer cell growth and augments proteasome inhibitor-induced effects. Secondly, specific knock-down of MKP-1 with siRNA significantly repressed cell viability by reduced cyclin D1 expression, and enhanced repression of cancer cell growth by proteasome inhibitors. Taken together, these results indicate that removing the unwanted (MKP-1-inducing) effects of bortezomib significantly improves the efficacy of proteasome inhibition in breast cancer cells. Thus, future development of drugs targeting MKP-1 offer promise of combination therapies with reduced toxicity and enhanced cell death in breast cancer.
Collapse
|
46
|
Wang Q, Shi S, He W, Padilla MT, Zhang L, Wang X, Zhang B, Lin Y. Retaining MKP1 expression and attenuating JNK-mediated apoptosis by RIP1 for cisplatin resistance through miR-940 inhibition. Oncotarget 2015; 5:1304-14. [PMID: 24675421 PMCID: PMC4012727 DOI: 10.18632/oncotarget.1798] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The elucidation of chemoresistance mechanisms is important to improve cancer patient survival. In this report, we investigated the role and mechanism through which receptor-interacting protein 1 (RIP1), a mediator in cell survival and death signaling, participates in cancer's response to chemotherapy. In lung cancer cells, knockdown of RIP1 substantially increased cisplatin-induced apoptotic cytotoxicity, which was associated with robust JNK activation. The expression of the JNK inactivating phosphatase, MKP1, was substantially reduced in RIP1 knockdown cells. Although MKP1 protein stability was not altered by RIP1 suppression, the synthesis rate of MKP1 was dramatically reduced in RIP1-suppressed cells. Furthermore, we found that the expression of miR-940 was substantially increased in RIP1 knockdown cells. Knockdown of miR-940 restored MKP1 expression and attenuated cisplatin-induced JNK activation and cytotoxicity. Importantly, ectopic expression of MKP1 effectively attenuated cisplatin-induced JNK activation and cytotoxicity. In addition, activation of the JNK upstream signaling kinase, MKK4, was also potentiated in RIP1 knockdown cells. Altogether, our results suggest that RIP1 contributes to cisplatin resistance by suppressing JNK activation that involves releasing miR-940-mediated inhibition of MKP1 and suppressing activation of MKK4. Intervention targeting the RIP1/miR-940/MKP1/JNK pathway may be used to sensitize platinum-based chemotherapy.
Collapse
Affiliation(s)
- Qiong Wang
- Laboratory of Molecular and Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education at Sichuan University, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
WANG DONG, HAN SHENG, PENG RUI, JIAO CHENYU, WANG XING, HAN ZEGUANG, LI XIANGCHENG. DUSP28 contributes to human hepatocellular carcinoma via regulation of the p38 MAPK signaling. Int J Oncol 2014; 45:2596-604. [DOI: 10.3892/ijo.2014.2653] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 08/30/2014] [Indexed: 11/06/2022] Open
|
48
|
Apostolou P, Toloudi M, Kourtidou E, Mimikakou G, Vlachou I, Chatziioannou M, Papasotiriou I. Use of the comet assay technique for quick and reliable prediction of in vitro response to chemotherapeutics in breast and colon cancer. ACTA ACUST UNITED AC 2014; 21:14. [PMID: 25984497 PMCID: PMC4389674 DOI: 10.1186/2241-5793-21-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 06/25/2014] [Indexed: 12/02/2022]
Abstract
Background Determination of response to chemotherapy is a major requirement of personalized medicine. Resistance, whether developed or native, critically affects a treatment’s success. Single Cell Gel lectrophoresis - also known as a comet assay - is used to detect DNA damage at the level of individual eukaryotic cells. We assessed the use of comet assays in determining response to chemotherapeutic drugs that are widely used in breast and colon cancer. Results We treated human breast and colon cancer cell lines with melphalan, cisplatin, mechlorethamine or doxorubicin, as monotherapies. Drug activities varied even in the same cancer types, further demonstrating the heterogeneity of different cancer types. Conclusion The comet assay technique can provide reliable and quick results with minimum requirements and is applicable to a wide variety of drugs.
Collapse
Affiliation(s)
| | - Maria Toloudi
- Research Genetic Cancer Centre Ltd (R.G.C.C. Ltd), Filotas, Florina Greece
| | - Eleni Kourtidou
- Research Genetic Cancer Centre Ltd (R.G.C.C. Ltd), Filotas, Florina Greece
| | - Georgia Mimikakou
- Research Genetic Cancer Centre Ltd (R.G.C.C. Ltd), Filotas, Florina Greece
| | - Ioanna Vlachou
- Research Genetic Cancer Centre Ltd (R.G.C.C. Ltd), Filotas, Florina Greece
| | | | | |
Collapse
|
49
|
Abstract
Mitogen-activated protein kinases (MAPKs) mediate a wide variety of cellular behaviors in response to extracellular stimuli. One of the main subgroups, the p38 MAP kinases, has been implicated in a wide range of complex biologic processes, such as cell proliferation, cell differentiation, cell death, cell migration, and invasion. Dysregulation of p38 MAPK levels in patients are associated with advanced stages and short survival in cancer patients (e.g., prostate, breast, bladder, liver, and lung cancer). p38 MAPK plays a dual role as a regulator of cell death, and it can either mediate cell survival or cell death depending not only on the type of stimulus but also in a cell type specific manner. In addition to modulating cell survival, an essential role of p38 MAPK in modulation of cell migration and invasion offers a distinct opportunity to target this pathway with respect to tumor metastasis. The specific function of p38 MAPK appears to depend not only on the cell type but also on the stimuli and/or the isoform that is activated. p38 MAPK signaling pathway is activated in response to diverse stimuli and mediates its function by components downstream of p38. Extrapolation of the knowledge gained from laboratory findings is essential to address the clinical significance of p38 MAPK signaling pathways. The goal of this review is to provide an overview on recent progress made in defining the functions of p38 MAPK pathways with respect to solid tumor biology and generate testable hypothesis with respect to the role of p38 MAPK as an attractive target for intervention of solid tumors.
Collapse
Affiliation(s)
- Hari K Koul
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center, Shreveport, LA, USA ; Feist-Weiller Cancer Center, Shreveport, LA, USA ; Veterans Administration Medical Center, Shreveport, LA, USA
| | - Mantu Pal
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center, Shreveport, LA, USA ; Veterans Administration Medical Center, Shreveport, LA, USA
| | - Sweaty Koul
- Feist-Weiller Cancer Center, Shreveport, LA, USA ; Department of Urology, LSU Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
50
|
Ríos P, Nunes-Xavier CE, Tabernero L, Köhn M, Pulido R. Dual-specificity phosphatases as molecular targets for inhibition in human disease. Antioxid Redox Signal 2014; 20:2251-73. [PMID: 24206177 DOI: 10.1089/ars.2013.5709] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
SIGNIFICANCE The dual-specificity phosphatases (DUSPs) constitute a heterogeneous group of cysteine-based protein tyrosine phosphatases, whose members exert a pivotal role in cell physiology by dephosphorylation of phosphoserine, phosphothreonine, and phosphotyrosine residues from proteins, as well as other non-proteinaceous substrates. RECENT ADVANCES A picture is emerging in which a selected group of DUSP enzymes display overexpression or hyperactivity that is associated with human disease, especially human cancer, making feasible targeted therapy approaches based on their inhibition. A panoply of molecular and functional studies on DUSPs have been performed in the previous years, and drug-discovery efforts are ongoing to develop specific and efficient DUSP enzyme inhibitors. This review summarizes the current status on inhibitory compounds targeting DUSPs that belong to the MAP kinase phosphatases-, small-sized atypical-, and phosphatases of regenerating liver subfamilies, whose inhibition could be beneficial for the prevention or mitigation of human disease. CRITICAL ISSUES Achieving specificity, potency, and bioavailability are the major challenges in the discovery of DUSP inhibitors for the clinics. Clinical validation of compounds or alternative inhibitory strategies of DUSP inhibition has yet to come. FUTURE DIRECTIONS Further work is required to understand the dual role of many DUSPs in human cancer, their function-structure properties, and to identify their physiologic substrates. This will help in the implementation of therapies based on DUSPs inhibition.
Collapse
Affiliation(s)
- Pablo Ríos
- 1 Genome Biology Unit, European Molecular Biology Laboratory , Heidelberg, Germany
| | | | | | | | | |
Collapse
|