1
|
Moghbeli M. MicroRNAs as the critical regulators of bone metastasis during prostate tumor progression. Int J Biol Macromol 2025; 309:142912. [PMID: 40203904 DOI: 10.1016/j.ijbiomac.2025.142912] [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: 03/01/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Prostate cancer (PCa) is the most prevalent cancer among men globally. Although, there are various therapeutic methods for the localized or advanced cancers, there is still a high rate of mortality among PCa patients that is mainly associated with bone metastasis in advanced tumors. There are few options available for treating bone metastasis in PCa, which only provide symptom relief without curing the disease. Therefore, it is crucial to evaluate the molecular mechanisms associated with bone metastasis of PCa cells to suggest the novel diagnostic and therapeutic approaches that could lower the morbidity and mortality rates in PCa patients. MicroRNAs (miRNAs) are involved in regulation of various pathophysiological processes such as tumor growth and osteoblasts/osteoclasts formation. Since, miRNA deregulation has been also frequently observed in PCa patients with bone metastasis, we discussed the role of miRNAs in bone metastasis during PCa progression. It has been reported that miRNAs mainly reduced the ability of PCa tumor cells for the bone metastasis through the regulation of WNT, NF-kB, PI3K/AKT, and TGF-β signaling pathways. They also affected the EMT process, transcription factors, and structural proteins to regulate the bone metastasis during PCa progression. This review paves the way to suggest the miRNAs as the reliable markers not only for the non-invasive early diagnosis, but also for the targeted therapy of PCa tumors with bone metastasis.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Kundu G, Ghasemi M, Yim S, Rohil A, Xin C, Ren L, Srivastava S, Akinfolarin A, Kumar S, Srivastava GP, Sabbisetti VS, Murugaiyan G, Ajay AK. STAT3 Protein-Protein Interaction Analysis Finds P300 as a Regulator of STAT3 and Histone 3 Lysine 27 Acetylation in Pericytes. Biomedicines 2024; 12:2102. [PMID: 39335615 PMCID: PMC11428717 DOI: 10.3390/biomedicines12092102] [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: 07/29/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Signal transducer and activator of transcription 3 (STAT3) is a member of the cytoplasmic inducible transcription factors and plays an important role in mediating signals from cytokines, chemokines, and growth factors. We and others have found that STAT3 directly regulates pro-fibrotic signaling in the kidney. The STAT3 protein-protein interaction plays an important role in activating its transcriptional activity. It is necessary to identify these interactions to investigate their function in kidney disease. Here, we investigated the protein-protein interaction among three species to find crucial interactions that can be targeted to alleviate kidney disease. METHOD In this study, we examined common protein-protein interactions leading to the activation or downregulation of STAT3 among three different species: humans (Homo sapiens), mice (Mus musculus), and rabbits (Oryctolagus cuniculus). Further, we chose to investigate the P300 and STAT3 interaction and performed studies of the activation of STAT3 using IL-6 and inhibition of the P300 by its specific inhibitor A-485 in pericytes. Next, we performed immunoprecipitation to confirm whether A-485 inhibits the binding of P300 to STAT3. RESULTS Using the STRING application from ExPASy, we found that six proteins, including PIAS3, JAK1, JAK2, EGFR, SRC, and EP300, showed highly confident interactions with STAT3 in humans, mice, and rabbits. We also found that IL-6 treatment increased the acetylation of STAT3 and increased histone 3 lysine acetylation (H3K27ac). Furthermore, we found that the disruption of STAT3 and P300 interaction by the P300 inhibitor A-485 decreased STAT3 acetylation and H3K27ac. Finally, we confirmed that the P300 inhibitor A-485 inhibited the binding of STAT3 with P300, which inhibited its transcriptional activity by reducing the expression of Ccnd1 (Cyclin D1). CONCLUSIONS Targeting the P300 protein interaction with STAT3 may alleviate STAT3-mediated fibrotic signaling in humans and other species.
Collapse
Affiliation(s)
- Gautam Kundu
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- US Military HIV Research Program (MHRP), Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Maryam Ghasemi
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Seungbin Yim
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Ayanna Rohil
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Cuiyan Xin
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Leo Ren
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | | | - Akinwande Akinfolarin
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Subodh Kumar
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Gyan P. Srivastava
- Department of Electrical Engineering & Computer Science, University of Missouri, Columbia, MO 65211, USA
| | - Venkata S. Sabbisetti
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Gopal Murugaiyan
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Amrendra K. Ajay
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Center for Polycystic Kidney Disease, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
3
|
Ma Z, Zhou F, Jin H, Wu X. Crosstalk between CXCL12/CXCR4/ACKR3 and the STAT3 Pathway. Cells 2024; 13:1027. [PMID: 38920657 PMCID: PMC11201928 DOI: 10.3390/cells13121027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
The reciprocal modulation between the CXCL12/CXCR4/ACKR3 axis and the STAT3 signaling pathway plays a crucial role in the progression of various diseases and neoplasms. Activation of the CXCL12/CXCR4/ACKR3 axis triggers the STAT3 pathway through multiple mechanisms, while the STAT3 pathway also regulates the expression of CXCL12. This review offers a thorough and systematic analysis of the reciprocal regulatory mechanisms between the CXCL12/CXCR4/ACKR3 signaling axis and the STAT3 signaling pathway in the context of diseases, particularly tumors. It explores the potential clinical applications in tumor treatment, highlighting possible therapeutic targets and novel strategies for targeted tumor therapy.
Collapse
Affiliation(s)
| | | | | | - Xiaoming Wu
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming 650500, China; (Z.M.); (F.Z.); (H.J.)
| |
Collapse
|
4
|
Zheng X, Wang L, Zhang Z, Tang H. The emerging roles of SUMOylation in pulmonary diseases. Mol Med 2023; 29:119. [PMID: 37670258 PMCID: PMC10478458 DOI: 10.1186/s10020-023-00719-1] [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: 06/20/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023] Open
Abstract
Small ubiquitin-like modifier mediated modification (SUMOylation) is a critical post-translational modification that has a broad spectrum of biological functions, including genome replication and repair, transcriptional regulation, protein stability, and cell cycle progression. Perturbation or deregulation of a SUMOylation and deSUMOylation status has emerged as a new pathophysiological feature of lung diseases. In this review, we highlighted the link between SUMO pathway and lung diseases, especially the sumoylated substrate such as C/EBPα in bronchopulmonary dysplasia (BDP), PPARγ in pneumonia, TFII-I in asthma, HDAC2 in chronic obstructive pulmonary disease (COPD), KLF15 in hypoxic pulmonary hypertension (HPH), SMAD3 in idiopathic pulmonary fibrosis (IPF), and YTHDF2 in cancer. By exploring the impact of SUMOylation in pulmonary diseases, we intend to shed light on its potential to inspire the development of innovative diagnostic and therapeutic strategies, holding promise for improving patient outcomes and overall respiratory health.
Collapse
Affiliation(s)
- Xuyang Zheng
- Department of pediatrics, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, P.R. China.
| | - Lingqiao Wang
- Department of pediatrics, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, P.R. China
| | - Zhen Zhang
- Department of Orthopedics Surgery, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 31000, Zhejiang, P.R. China
| | - Huifang Tang
- Department of Pharmacology, Zhejiang Respiratory Drugs Research Laboratory, School of Basic Medicial Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, P.R. China.
| |
Collapse
|
5
|
Abolghasemi S, Bikhof Torbati M, Pakzad P, Ghafouri-Fard S. Gene expression analysis of SOCS, STAT and PIAS genes in lung cancer patients. Pathol Res Pract 2023; 249:154760. [PMID: 37586215 DOI: 10.1016/j.prp.2023.154760] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/05/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023]
Abstract
One of the most devastating diseases with the highest prevalence and mortality rate worldwide is lung cancer. Non-small cell lung cancer (NSCLC) is the subtype of lung cancer in 85% of cases. In this work, the expression levels of the STAT, SOCS and PIAS family genes involved in angiogenesis, proliferation and differentiation were examined. Using QRT-PCR technique, the expression level of STAT3 gene was assessed and tumor tissue samples had higher expression than normal tissue. In addition, the histological grade of adenocarcinoma was associated with the increase in STAT3 gene expression. The expression of the SOCS1 and SOCS2 genes in tumors was measured to be 0.58-fold and 0.36-fold lower than in healthy samples adjacent to the tumor, but this reduction in expression was not significant. In addition, when examining the relationship between the expression of SOCS1 and 2 and the clinical features of tumor samples, there was a significant decrease in the expression of the SOCS1 and 2 genes in the adenocarcinoma subtype. Compared to neighboring tumor samples, the expression of PIAS1 in the tumors was not different with controls. Our research revealed that tissue samples from adenocarcinoma had higher levels of STAT3 expression. Taken together, the mentioned genes can be suggested as possible targets for further studies in NSCLC.
Collapse
Affiliation(s)
- Solmaz Abolghasemi
- College of Biology Sciences, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Maryam Bikhof Torbati
- Department of Biology, Yadegar-e-Imam Khomeini(RAH) Shahr-e-Rey Branch, Islamic Azad University, Tehran, Iran
| | - Parviz Pakzad
- Department of Microbiology, School of Biology Sciences, North Branch, Islamic Azad University, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Liu H, Zhang J, Xue Z, Chang M, Feng X, Cai Y, Bai L, Wang W, Liu E, Zhao S, Wang R. Deficiency of protein inhibitor of activated STAT3 exacerbates atherosclerosis by modulating VSMC phenotypic switching. Atherosclerosis 2023; 380:117195. [PMID: 37586220 DOI: 10.1016/j.atherosclerosis.2023.117195] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 07/22/2023] [Accepted: 07/26/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND AND AIMS Phenotypic switching of vascular smooth muscle cells (VSMCs) plays an essential role in the development of atherosclerosis. Protein inhibitor of activated STAT (Pias) regulates VSMCs phenotype via acting as sumo E3 ligase to promote protein sumoylation. Our previous study indicated that Pias3 expression decreased in atherosclerotic lesions. Therefore, this study aimed to explore the role of Pias3 on VSMCs phenotype switching during atherosclerosis. METHODS ApoE-/- and ApoE-/-Pias3-/- double-deficient mice were fed with high-fat/high-cholesterol diet to induce atherosclerosis. Aorta tissues and primary VSMCs were collected to assess plaque formation and VSMCs phenotype. In vitro, Pias3 was overexpressed in A7r5, a VSMCs cell line, by transfection with Pias3 plasmid. Real-time quantitative PCR, immunoblotting, immunoprecipitation, were used to analyze the effect of Pias3 on VSMCs phenotypic switching. RESULTS Pias3 deficiency significantly exacerbated atherosclerotic plaque formation and promoted VSMCs phenotypic switching to a synthetic state within lesion. In vitro, overexpressing Pias3 in VSMCs increased the expression of contractile markers (myosin heavy chain 11, calponin 1), while it decreased the level of synthetic marker (vimentin). Additionally, Pias3 overexpression blocked PDGF-BB-induced VSMCs proliferation and migration. Immunoprecipitation and mass spectrometry results showed that Pias3 enhanced sumoylation and ubiquitination of vimentin, and shortened its half-life. Moreover, the ubiquitination level of vimentin was impaired by 2-D08, a sumoylation inhibitor. This suggests that Pias3 might accelerate the ubiquitination-degradation of vimentin by promoting its sumoylation. CONCLUSIONS These results indicate that Pias3 might ameliorate atherosclerosis progression by suppressing VSMCs phenotypic switching and reducing vimentin protein stability.
Collapse
Affiliation(s)
- Haole Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jingyi Zhang
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ziyang Xue
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Mingke Chang
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xinxin Feng
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yifan Cai
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Liang Bai
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China; Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Weirong Wang
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China; Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Enqi Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China; Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Sihai Zhao
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Rong Wang
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
7
|
Faida P, Attiogbe MKI, Majeed U, Zhao J, Qu L, Fan D. Lung cancer treatment potential and limits associated with the STAT family of transcription factors. Cell Signal 2023:110797. [PMID: 37423343 DOI: 10.1016/j.cellsig.2023.110797] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/19/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023]
Abstract
Lung cancer is one of the mortal cancers and the leading cause of cancer-related mortality, with a cancer survival rate of fewer than 5% in developing nations. This low survival rate can be linked to things like late-stage detection, quick postoperative recurrences in patients receiving therapy, and chemoresistance developing against various lung cancer treatments. Signal transducer and activator of transcription (STAT) family of transcription factors are involved in lung cancer cell proliferation, metastasis, immunological control, and treatment resistance. By interacting with specific DNA sequences, STAT proteins trigger the production of particular genes, which in turn result in adaptive and incredibly specific biological responses. In the human genome, seven STAT proteins have been discovered (STAT1 to STAT6, including STAT5a and STAT5b). Many external signaling proteins can activate unphosphorylated STATs (uSTATs), which are found inactively in the cytoplasm. When STAT proteins are activated, they can increase the transcription of several target genes, which leads to unchecked cellular proliferation, anti-apoptotic reactions, and angiogenesis. The effects of STAT transcription factors on lung cancer are variable; some are either pro- or anti-tumorigenic, while others maintain dual, context-dependent activities. Here, we give a succinct summary of the various functions that each member of the STAT family plays in lung cancer and go into more detail about the advantages and disadvantages of pharmacologically targeting STAT proteins and their upstream activators in the context of lung cancer treatment.
Collapse
Affiliation(s)
- Paison Faida
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China
| | - Mawusse K I Attiogbe
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Usman Majeed
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China
| | - Jing Zhao
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China
| | - Linlin Qu
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
8
|
Clowers MJ, Moghaddam SJ. Cell Type-Specific Roles of STAT3 Signaling in the Pathogenesis and Progression of K-ras Mutant Lung Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14071785. [PMID: 35406557 PMCID: PMC8997152 DOI: 10.3390/cancers14071785] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Lung adenocarcinomas with mutations in the K-ras gene are hard to target pharmacologically and highly lethal. As a result, there is a need to identify other therapeutic targets that influence K-ras oncogenesis. One contender is STAT3, a transcription factor that is associated with K-ras mutations and aids tumor development and progression through tumor cell intrinsic and extrinsic mechanisms. In this review, we summarize the lung epithelial and infiltrating immune cells that express STAT3, the roles of STAT3 in K-ras mutant lung adenocarcinoma, and therapies that may be able to target STAT3. Abstract Worldwide, lung cancer, particularly K-ras mutant lung adenocarcinoma (KM-LUAD), is the leading cause of cancer mortality because of its high incidence and low cure rate. To treat and prevent KM-LUAD, there is an urgent unmet need for alternative strategies targeting downstream effectors of K-ras and/or its cooperating pathways. Tumor-promoting inflammation, an enabling hallmark of cancer, strongly participates in the development and progression of KM-LUAD. However, our knowledge of the dynamic inflammatory mechanisms, immunomodulatory pathways, and cell-specific molecular signals mediating K-ras-induced lung tumorigenesis is substantially deficient. Nevertheless, within this signaling complexity, an inflammatory pathway is emerging as a druggable target: signal transducer and activator of transcription 3 (STAT3). Here, we review the cell type-specific functions of STAT3 in the pathogenesis and progression of KM-LUAD that could serve as a new target for personalized preventive and therapeutic intervention for this intractable form of lung cancer.
Collapse
Affiliation(s)
- Michael J. Clowers
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
9
|
Li C, Wang R, Zhang Y, Hu C, Ma Q. PIAS3 suppresses damage in an Alzheimer's disease cell model by inducing the STAT3-associated STAT3/Nestin/Nrf2/HO-1 pathway. Mol Med 2021; 27:150. [PMID: 34837964 PMCID: PMC8626961 DOI: 10.1186/s10020-021-00410-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 11/09/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD), the most common form of dementia, is caused by the degeneration of the central nervous system (CNS). A previous study reported that signal transducer and activator of transcription 3 (STAT3) is activated during AD development; nonetheless, the related mechanism remains unknown. Thus, this study used a cell model to explore whether and how the protein inhibitor of activated STAT3 (PIAS3) is involved in AD development. METHODS Cerebrospinal fluid (CSF) specimens of 30 patients with AD and 10 normal participants were included in this study. SH-SY5Y cells were used to constructed AD model. Relevant indices were then detected and analyzed. RESULTS The results showed that compared with the control group, PIAS3 expression was substantially decreased in patients with AD and amyloid beta (Aβ)-treated SH-SY5Y cells. PIAS3 overexpression was able to reverse the detrimental effects of Aβ treatment on cell survival and growth. Further, it could also ameliorate apoptosis and oxidative stress in Aβ-treated SH-SY5Y cells. Additionally, PIAS3 was shown to reduce the activated form of STAT3 and increase the activity of the downstream Nestin/nuclear factor erythroid 2-related factor/heme oxygenase-1 pathway. CONCLUSIONS STAT3 reactivation by colivelin treatment negated the influence of PIAS3 on the survival, growth, apoptosis, and oxidative stress of Aβ-treated SH-SY5Y cells.
Collapse
Affiliation(s)
- Chen Li
- Department of Geriatrics Neurology, the Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Ruili Wang
- Department of Geriatrics Neurology, the Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Youyou Zhang
- Department of Geriatrics Neurology, the Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Chunting Hu
- Department of Geriatrics Neurology, the Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Qiaoya Ma
- Department of Geriatrics Neurology, the Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 Shaanxi People’s Republic of China
| |
Collapse
|
10
|
Sun H, Li Y, Quan X, Chen N, Jin X, Jin W, Jin Y, Shen X. PIAS3/SOCS1-STAT3 axis responses to oxidative stress in hepatocellular cancer cells. Am J Transl Res 2021; 13:12395-12409. [PMID: 34956461 PMCID: PMC8661178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/23/2021] [Indexed: 06/14/2023]
Abstract
The participation of STAT3 and its upstream inhibitors, PIAS3 and SOCS1, in the oxidative response of hepatocellular carcinoma (HCC) cells was uncertain. Here, the expression of PIAS3 and SOCS1 in HCC tissues and cell lines was explored, and we sought to determine whether oxidative stress epigenetically regulated PIAS3 and SOCS1 expression and STAT3 activation in HCC cells. The expression of PIAS3 and SOCS1 was markedly decreased in HCC cell lines and tissues compared to normal hepatic cells and tissues. In HCC patients, low PIAS3 and SOCS1 expression were associated with poor survival. Oxidative stress induced by H2O2 in HepG2 cells was indicated by low antioxidant levels and high protein carbonyl content. Moreover, oxidative stress in HepG2 cells contributed to reduced proliferation but increased apoptosis, migration, and invasion capacity, which might be counteracted by antioxidants, such as tocopheryl acetate (TA). PIAS3 and SOCS1 expression was markedly decreased, while STAT3 was activated in HepG2 cells in response to H2O2 exposure. Co-treatment with antioxidant TA effectively increased the expression of PIAS3 and SOCS1, but it dephosphorylated STAT3 in H2O2-treated cells. PIAS1 or SOCS1 overexpression in HepG2 cells after H2O2 treatment restored cell viability and anti-oxidative responses and decreased apoptosis, migration, and invasion ability, and dephosphorylated STAT3 levels. Co-administration of the STAT3 activator, colivelin, partially abolished the effect of PIAS3 and SOCS1 overexpression in these processes. Therefore, oxidative stress in HCC cells may improve their migration and reduce proliferation through STAT3 activation through the repression of PIAS3 and SOCS1 expression.
Collapse
Affiliation(s)
- Honghua Sun
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Yanglong Li
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Xianglan Quan
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Ning Chen
- Department of Infection Disease, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Xinglin Jin
- Department of General Surgert, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Wenbiao Jin
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Yongmin Jin
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| | - Xionghu Shen
- Department of Oncology, Affiliated Hospital of Yanbian UniversityYanji, Jilin Province, People’s Republic of China
| |
Collapse
|
11
|
Kukkula A, Ojala VK, Mendez LM, Sistonen L, Elenius K, Sundvall M. Therapeutic Potential of Targeting the SUMO Pathway in Cancer. Cancers (Basel) 2021; 13:4402. [PMID: 34503213 PMCID: PMC8431684 DOI: 10.3390/cancers13174402] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 02/07/2023] Open
Abstract
SUMOylation is a dynamic and reversible post-translational modification, characterized more than 20 years ago, that regulates protein function at multiple levels. Key oncoproteins and tumor suppressors are SUMO substrates. In addition to alterations in SUMO pathway activity due to conditions typically present in cancer, such as hypoxia, the SUMO machinery components are deregulated at the genomic level in cancer. The delicate balance between SUMOylation and deSUMOylation is regulated by SENP enzymes possessing SUMO-deconjugation activity. Dysregulation of SUMO machinery components can disrupt the balance of SUMOylation, contributing to the tumorigenesis and drug resistance of various cancers in a context-dependent manner. Many molecular mechanisms relevant to the pathogenesis of specific cancers involve SUMO, highlighting the potential relevance of SUMO machinery components as therapeutic targets. Recent advances in the development of inhibitors targeting SUMOylation and deSUMOylation permit evaluation of the therapeutic potential of targeting the SUMO pathway in cancer. Finally, the first drug inhibiting SUMO pathway, TAK-981, is currently also being evaluated in clinical trials in cancer patients. Intriguingly, the inhibition of SUMOylation may also have the potential to activate the anti-tumor immune response. Here, we comprehensively and systematically review the recent developments in understanding the role of SUMOylation in cancer and specifically focus on elaborating the scientific rationale of targeting the SUMO pathway in different cancers.
Collapse
Affiliation(s)
- Antti Kukkula
- Cancer Research Unit, FICAN West Cancer Center Laboratory, Institute of Biomedicine, Turku University Hospital, University of Turku, FI-20520 Turku, Finland; (A.K.); (V.K.O.); (K.E.)
| | - Veera K. Ojala
- Cancer Research Unit, FICAN West Cancer Center Laboratory, Institute of Biomedicine, Turku University Hospital, University of Turku, FI-20520 Turku, Finland; (A.K.); (V.K.O.); (K.E.)
- Turku Doctoral Programme of Molecular Medicine, University of Turku, FI-20520 Turku, Finland
- Medicity Research Laboratories, University of Turku, FI-20520 Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland;
| | - Lourdes M. Mendez
- Beth Israel Deaconess Cancer Center, Beth Israel Deaconess Medical Center, Department of Medicine and Pathology, Cancer Research Institute, Harvard Medical School, Boston, MA 02115, USA;
| | - Lea Sistonen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland;
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, FI-20520 Turku, Finland
| | - Klaus Elenius
- Cancer Research Unit, FICAN West Cancer Center Laboratory, Institute of Biomedicine, Turku University Hospital, University of Turku, FI-20520 Turku, Finland; (A.K.); (V.K.O.); (K.E.)
- Medicity Research Laboratories, University of Turku, FI-20520 Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland;
- Department of Oncology, Turku University Hospital, FI-20521 Turku, Finland
| | - Maria Sundvall
- Cancer Research Unit, FICAN West Cancer Center Laboratory, Institute of Biomedicine, Turku University Hospital, University of Turku, FI-20520 Turku, Finland; (A.K.); (V.K.O.); (K.E.)
- Department of Oncology, Turku University Hospital, FI-20521 Turku, Finland
| |
Collapse
|
12
|
Wang J, Fu J, Xu C, Jia R, Zhang X, Zhao S. Circ_ZFP644 attenuates caerulein-induced inflammatory injury in rat pancreatic acinar cells by modulating miR-106b/Pias3 axis. Exp Mol Pathol 2021; 121:104644. [PMID: 33945806 DOI: 10.1016/j.yexmp.2021.104644] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 03/30/2021] [Accepted: 04/29/2021] [Indexed: 11/16/2022]
Abstract
(AP) is a kind of inflammatory misorder existing in pancreas. Non-coding RNAs (ncRNAs) have been reported to play important roles in development of AP. The current study was designed to explore the role of circular RNA zinc finger protein 644 (circRNA circ_ZFP644) in caerulein-induced AR42J cells. AP model in vitro was established by exposure of rat pancreatic acinar AR42J cells to caerulein. Amylase activity was measured using a kit. Enzyme-linked immunosorbent assay (ELISA) was performed to examine the levels of several inflammatory factors. The expression of circ_ZFP644, microRNA (miR)-106b and protein inhibitor of activated STAT 3 (Pias3) was detected by quantitative real-time PCR (qRT-PCR) or western blot assay. And flow cytometry was employed to monitor cell apoptosis. Western blot assay was also conducted to analyze the expression of apoptosis-related proteins. The association among circ_ZFP644, miR-106b and Pias3 was validated by dual-luciferase reporter assay. Caerulein treatment activated amylase activity and promoted the secretion of inflammatory cytokines in AR42J cells. Circ_ZFP644 and Pias3 were downregulated, but miR-106b was upregulated in caerulein-induced AR42J cells. Enforced expression of circ_ZFP644 or miR-106b inhibition could reduce amylase activity and inflammatory cytokine secretion, while promote apoptosis in caerulein-induced AR42J cells, which was almost reversed by Pias3 knockdown. Circ_ZFP644 targeted miR-106b to upregulate Pias3 expression. Circ_ZFP644 might exert its anti-inflammation and pro-apoptosis roles in caerulein-induced AR42J cells by regulating miR-106b/Pias3 axis.
Collapse
Affiliation(s)
- Jing Wang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jindong Fu
- Department of Gastroenterology, Rizhao People's Hospital, Rizhao, Shandong, China
| | - Changqin Xu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ruzhen Jia
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaohua Zhang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shulei Zhao
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
13
|
ROR2 suppresses metastasis of prostate cancer via regulation of miR-199a-5p-PIAS3-AKT2 signaling axis. Cell Death Dis 2020; 11:376. [PMID: 32415173 PMCID: PMC7228945 DOI: 10.1038/s41419-020-2587-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 12/18/2022]
Abstract
Bones are the most common metastatic sites for prostate cancer (PCa). Receptor tyrosine kinase-like orphan receptor 2 (ROR2), a noncanonical Wnt receptor, plays crucial roles in skeletal morphogenesis, osteoblast differentiation, and bone formation. The role of ROR2 in PCa metastasis is unclear. We analyzed online datasets from Oncomine as well as using IHC staining on tissue array to determine the relationship between ROR2 expression level and disease outcome of PCa. To investigate how ROR2 regulates migration and invasion of PCa cells, we performed transwell assay and orthotopic xenograft model in nude mice. We then applied the Micro-Western Array (MWA), a high-throughput western blotting platform to analyze the downstream signaling pathways being regulated by ROR2. Compared with nonmalignant PZ-HPV-7 and RWPE-1 cells, PCa cell lines express lower level of ROR2 protein. Constitutive expression of ROR2 in PC-3, DU-145, or C4-2B PCa cells significantly suppressed the cell migration, invasion, and epithelial-mesenchymal transition (EMT) proteins. MWA, western blotting, and microRNA analysis showed that elevation of ROR2 suppressed the expression of miR-199a-5p, which in turn increased the expression of PIAS3. The upregulation of PIAS3 then decreased AKT2 and the phosphorylation of AKT, resulting in the inhibition of migration and invasion of PCa cells both in vitro and in orthotopic xenograft mice model. IHC staining of tissue array and Oncomine datasets analysis indicated that the gene and protein level of ROR2 is much lower in metastatic prostate tumors as compared with primary tumors or adjacent normal prostate tissues. Low level of ROR2 correlated to poor survival and high recurrent frequency in PCa patients. In conclusion, we discovered that ROR2 suppresses PCa metastasis via regulation of PIAS3-PI3K-AKT2 signaling axis.
Collapse
|
14
|
Khan M, Maryam A, Saleem MZ, Shakir HA, Qazi JI, Li Y, Ma T. Brevilin A induces ROS-dependent apoptosis and suppresses STAT3 activation by direct binding in human lung cancer cells. J Cancer 2020; 11:3725-3735. [PMID: 32328177 PMCID: PMC7171504 DOI: 10.7150/jca.40983] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/06/2020] [Indexed: 12/30/2022] Open
Abstract
Sesquiterpene lactones have been shown to be promising leads for anticancer drug development. Brevilin A (BLN-A), a sesquiterpene lactone compound of Centipeda minima has been shown to exhibit anticancer effects against various cancer cells. However, the anticancer mechanism and cellular targets of BLN-A remain elusive. Here in this study, BLN-A inhibits proliferation and induces cell morphological changes in A549 and NCI-H1650 non-small cell lung cancer cells in a dose-dependent manner. Moreover, BLN-A increased ROS generation and bax/bcl-2 ratio while decreased intracellular glutathione (GSH), and mitochondrial membrane potential which resulted in induction of apoptosis as evident by annexin-V/FITC staining, caspase-3 activation and PARP cleavage. Supplementation of cells with NAC (ROS Scavenger) effectively protected the cells from BLN-A-induced apoptosis. Finally, BLN-A inhibited constitutive as well as IL-6- and EGF-induced STAT3 activation at Tyr705. Using molecular docking and SPR analyses, we found that BLN-A directly binds with STAT3 and thereby inhibits its activation. Knocking down of STAT3 by stable transfection with shRNA suppressed growth and augmented cytotoxicity of BLN-A, indicating the key role of STAT3 in BLN-A-mediated apoptosis. Cumulative findings suggest that BLN-A is a promising lead structure for developing it into a potent STAT3 inhibitor and therapeutic agent against NSCLC as well.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore-54590, Pakistan
| | - Amara Maryam
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore-54590, Pakistan
| | | | - Hafiz Abdullah Shakir
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore-54590, Pakistan
| | - Javed Iqbal Qazi
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore-54590, Pakistan
| | - Yongming Li
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Tonghui Ma
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| |
Collapse
|
15
|
Owonikoko TK, Harvey RD, Carthon B, Chen Z, Lewis C, Collins H, Zhang C, Lawson DH, Alese OB, Bilen MA, Sica GL, Steuer CE, Shaib WL, Wu C, Harris WB, Akce M, Kudchagkar RR, El-Rayes BF, Lonial S, Ramalingam SS, Khuri FR. A Phase I Study of Safety, Pharmacokinetics, and Pharmacodynamics of Concurrent Everolimus and Buparlisib Treatment in Advanced Solid Tumors. Clin Cancer Res 2020; 26:2497-2505. [PMID: 32005746 DOI: 10.1158/1078-0432.ccr-19-2697] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/19/2019] [Accepted: 01/27/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Concurrent inhibition of mTOR and PI3K led to improved efficacy in preclinical models and provided the rationale for this phase I study of everolimus and buparlisib (BKM120) in patients with advanced solid tumor. PATIENTS AND METHODS We used the Bayesian Escalation with Overdose Control design to test escalating doses of everolimus (5 or 10 mg) and buparlisib (20, 40, 60, 80, and 100 mg) in eligible patients. Pharmacokinetic assessment was conducted using blood samples collected on cycle 1, days 8 and 15. Pharmacodynamic impact on mTOR/PI3K pathway modulation evaluated in paired skin biopsies collected at baseline and end of cycle 1. RESULTS We enrolled 43 patients, median age of 63 (range, 39-78) years; 25 (58.1%) females, 35 (81.4%) Caucasians, and 8 (18.6%) Blacks. The most frequent toxicities were hyperglycemia, diarrhea, nausea, fatigue, and aspartate aminotransferase elevation. Dose-limiting toxicities observed in 7 patients were fatigue (3), hyperglycemia (2), mucositis (1), acute kidney injury (1), and urinary tract infection (1). The recommended phase II dose (RP2D) for the combination was established as everolimus (5 mg) and buparlisib (60 mg). The best response in 27 evaluable patients was progressive disease and stable disease in 3 (11%) and 24 (89%), respectively. The median progression-free survival and overall survival were 2.7 (1.8-4.2) and 9 (6.4-13.2) months. Steady-state pharmacokinetic analysis showed dose-normalized maximum concentrations and AUC values for everolimus and buparlisib in combination to be comparable with single-agent pharmacokinetic. CONCLUSIONS The combination of everolimus and buparlisib is safe and well-tolerated at the RP2D of 5 and 60 mg on a continuous daily schedule.
Collapse
Affiliation(s)
- Taofeek K Owonikoko
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia. .,Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - R Donald Harvey
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia.,Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Bradley Carthon
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia
| | - Zhengjia Chen
- Winship Cancer Institute of Emory University, Atlanta, Georgia.,Department of Statistics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Colleen Lewis
- Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Hanna Collins
- Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Chao Zhang
- Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - David H Lawson
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia
| | - Olatunji B Alese
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia
| | - Mehmet Asim Bilen
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia
| | - Gabriel L Sica
- Department of Statistics, Rollins School of Public Health, Emory University, Atlanta, Georgia.,Department of Pathology, Emory University, Atlanta, Georgia
| | - Conor E Steuer
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia
| | - Walid L Shaib
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia
| | - Christina Wu
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia
| | - Wayne B Harris
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia
| | - Mehmet Akce
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia
| | - Ragini R Kudchagkar
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia
| | - Bassel F El-Rayes
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia.,Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Sagar Lonial
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia.,Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Suresh S Ramalingam
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia.,Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Fadlo Raja Khuri
- Department of Hematology & Medical Oncology, Emory University, Atlanta, Georgia.,American University of Beirut, Beirut, Lebanon
| |
Collapse
|
16
|
Inhibition of JAK2/STAT3 signaling pathway protects mice from the DDP-induced acute kidney injury in lung cancer. Inflamm Res 2019; 68:751-760. [DOI: 10.1007/s00011-019-01258-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/20/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022] Open
|
17
|
Wu M, Song D, Li H, Yang Y, Ma X, Deng S, Ren C, Shu X. Negative regulators of STAT3 signaling pathway in cancers. Cancer Manag Res 2019; 11:4957-4969. [PMID: 31213912 PMCID: PMC6549392 DOI: 10.2147/cmar.s206175] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/17/2019] [Indexed: 12/19/2022] Open
Abstract
STAT3 is the most ubiquitous member of the STAT family and involved in many biological processes, such as cell proliferation, differentiation, and apoptosis. Mounting evidence has revealed that STAT3 is aberrantly activated in many malignant tumors and plays a critical role in cancer progression. STAT3 is usually regarded as an effective molecular target for cancer treatment, and abolishing the STAT3 activity may diminish tumor growth and metastasis. Recent studies have shown that negative regulators of STAT3 signaling such as PIAS, SOCS, and PTP, can effectively retard tumor progression. However, PIAS, SOCS, and PTP have also been reported to correlate with tumor malignancy, and their biological function in tumorigenesis and antitumor therapy are somewhat controversial. In this review, we summarize actual knowledge on the negative regulators of STAT3 in tumors, and focus on the potential role of PIAS, SOCS, and PTP in cancer treatment. Furthermore, we also outline the STAT3 inhibitors that have entered clinical trials. Targeting STAT3 seems to be a promising strategy in cancer therapy.
Collapse
Affiliation(s)
- Moli Wu
- College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China.,College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Danyang Song
- College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Hui Li
- College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Yang Yang
- College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Xiaodong Ma
- College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Sa Deng
- College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Changle Ren
- Surgery Department of Dalian Municipal Central Hospital, Dalian Medical University, Dalian 116033, People's Republic of China
| | - Xiaohong Shu
- College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| |
Collapse
|
18
|
Qu D, Yang Y, Huang X. miR-199a-5p promotes proliferation and metastasis and epithelial-mesenchymal transition through targeting PIAS3 in cervical carcinoma. J Cell Biochem 2019; 120:13562-13572. [PMID: 30937952 DOI: 10.1002/jcb.28631] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/29/2019] [Accepted: 02/04/2019] [Indexed: 12/13/2022]
Abstract
Cervical carcinoma is the second most frequent gynecological malignancies in females worldwide. The objective of this study was to investigate the role of miR-199a-5p and protein inhibitor of activated signal transducer and activators of transcription 3 (PIAS3) in cervical carcinoma. Quantitative reverse transcription polymerase chain reaction was utilized to detect miR-199a-5p and PIAS3 expression in cervical carcinoma tissues and cells. 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide Kit, transwell assay were used to explore the biological functions of miR-199a-5p in cervical carcinoma. Western blot analysis was applied to determine the expression level of epithelial-mesenchymal transition (EMT)-associated proteins and PIAS3 expression. The relationship between miR-199a-5p and PIAS3 was verified by luciferase activity reporter assay. We found that miR-199a-5p was upregulated in cervical carcinoma tissues and cell lines, and overexpression of miR-199a-5p promoted cell proliferation and metastasis in cervical carcinoma. In addition, Western blot analysis indicated that the enforced upregulation of miR-199a-5p enhanced mesenchymal markers vimentin and N-cadherin expressions, whereas reduced epithelial marker E-cadherin expressions. miR-199a-5p directly targeted PIAS3 and negatively regulated PIAS3 level in cervical carcinoma cells. And upregulation of PIAS3 reversed the effects of miR-199a-5p in cervical carcinoma. Collectively, our data provide evidence for miR-199a-5p function in cervical carcinoma growth, EMT, and metastasis; it may be act as a therapeutic strategy target for patients with cervical carcinoma.
Collapse
Affiliation(s)
- Dacheng Qu
- Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, People's Republic of China
| | - Yamei Yang
- Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, People's Republic of China
| | - Xiaoping Huang
- Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, People's Republic of China
| |
Collapse
|
19
|
Jiao J, Zhang R, Li Z, Yin Y, Fang X, Ding X, Cai Y, Yang S, Mu H, Zong D, Chen Y, Zhang Y, Zou J, Shao J, Huang Z. Nuclear Smad6 promotes gliomagenesis by negatively regulating PIAS3-mediated STAT3 inhibition. Nat Commun 2018; 9:2504. [PMID: 29950561 PMCID: PMC6021382 DOI: 10.1038/s41467-018-04936-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 06/06/2018] [Indexed: 12/11/2022] Open
Abstract
To date, the molecular mechanism underlying constitutive signal transducer and activator of transcription 3 (STAT3) activation in gliomas is largely unclear. In this study, we report that Smad6 is overexpressed in nuclei of glioma cells, which correlates with poor patient survival and regulates STAT3 activity via negatively regulating the Protein Inhibitors of Activated STAT3 (PIAS3). Mechanically, Smad6 interacts directly with PIAS3, and this interaction is mediated through the Mad homology 2 (MH2) domain of Smad6 and the Ring domain of PIAS3. Smad6 recruits Smurf1 to facilitate PIAS3 ubiquitination and degradation, which also depends on the MH2 domain and the PY motif of Smad6. Consequently, Smad6 reduces PIAS3-mediated STAT3 inhibition and promotes glioma cell growth and stem-like cell initiation. Moreover, the Smad6 MH2 transducible protein restores PIAS3 expression and subsequently reduces gliomagenesis. Collectively, we conclude that nuclear-Smad6 enhances glioma development by inducing PIAS3 degradation and subsequent STAT3 activity upregulation.
Collapse
Affiliation(s)
- Jiantong Jiao
- Center of Clinical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
- Wuxi Institute of Translational Medicine, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Rui Zhang
- Center of Clinical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Zheng Li
- Center of Clinical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
- Wuxi Institute of Translational Medicine, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Ying Yin
- Center of Clinical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
- Wuxi Institute of Translational Medicine, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Xiangming Fang
- Department of Radiology, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Xiaopeng Ding
- Center of Clinical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Ying Cai
- Department of Pathology, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Shudong Yang
- Department of Pathology, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Huijun Mu
- Center of Clinical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
- Wuxi Institute of Translational Medicine, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Da Zong
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Yuexin Chen
- Center of Clinical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
- Wuxi Institute of Translational Medicine, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Yansong Zhang
- Department of Neurosurgery, Nanjing Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jian Zou
- Center of Clinical Research, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China.
- Wuxi Institute of Translational Medicine, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China.
| | - Junfei Shao
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China.
- Wuxi Institute of Translational Medicine, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214023, China.
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China.
| |
Collapse
|
20
|
STAT3 Interactors as Potential Therapeutic Targets for Cancer Treatment. Int J Mol Sci 2018; 19:ijms19061787. [PMID: 29914167 PMCID: PMC6032216 DOI: 10.3390/ijms19061787] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 02/07/2023] Open
Abstract
Signal transducers and activators of transcription (STATs) mediate essential signaling pathways in different biological processes, including immune responses, hematopoiesis, and neurogenesis. Among the STAT members, STAT3 plays crucial roles in cell proliferation, survival, and differentiation. While STAT3 activation is transient in physiological conditions, STAT3 becomes persistently activated in a high percentage of solid and hematopoietic malignancies (e.g., melanoma, multiple myeloma, breast, prostate, ovarian, and colon cancers), thus contributing to malignant transformation and progression. This makes STAT3 an attractive therapeutic target for cancers. Initial strategies aimed at inhibiting STAT3 functions have focused on blocking the action of its activating kinases or sequestering its DNA binding ability. More recently, the diffusion of proteomic-based techniques, which have allowed for the identification and characterization of novel STAT3-interacting proteins able to modulate STAT3 activity via its subcellular localization, interact with upstream kinases, and recruit transcriptional machinery, has raised the possibility to target such cofactors to specifically restrain STAT3 oncogenic functions. In this article, we summarize the available data about the function of STAT3 interactors in malignant cells and discuss their role as potential therapeutic targets for cancer treatment.
Collapse
|
21
|
Saini U, Suarez AA, Naidu S, Wallbillich JJ, Bixel K, Wanner RA, Bice J, Kladney RD, Lester J, Karlan BY, Goodfellow PJ, Cohn DE, Selvendiran K. STAT3/PIAS3 Levels Serve as "Early Signature" Genes in the Development of High-Grade Serous Carcinoma from the Fallopian Tube. Cancer Res 2018; 78:1739-1750. [PMID: 29339537 DOI: 10.1158/0008-5472.can-17-1671] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 11/13/2017] [Accepted: 01/10/2018] [Indexed: 01/23/2023]
Abstract
The initial molecular events that lead to malignant transformation of the fimbria of the fallopian tube (FT) through high-grade serous ovarian carcinoma (HGSC) remain poorly understood. In this study, we report that increased expression of signal transducer and activator of transcription 3 (pSTAT3 Tyr705) and suppression or loss of protein inhibitor of activated STAT3 (PIAS3) in FT likely drive HGSC. We evaluated human tissues-benign normal FT, tubal-peritoneal junction (TPJ), p53 signature FT tissue, tubal intraepithelial lesion in transition (TILT), serous tubal intraepithelial carcinoma (STIC) without ovarian cancer, and HGSC for expression of STAT3/PIAS3 (compared with their known TP53 signature) and their target proliferation genes. We observed constitutive activation of STAT3 and low levels or loss of PIAS3 in the TPJ, p53 signature, TILT, and STIC through advanced stage IV (HGSC) tissues. Elevated expression of pSTAT3 Tyr705 and decreased levels of PIAS3 appeared as early as TPJ and the trend continued until very advanced stage HGSC (compared with high PIAS3 and low pSTAT3 expression in normal benign FT). Exogenous expression of STAT3 in FT cells mediated translocation of pSTAT3 and c-Myc into the nucleus. In vivo experiments demonstrated that overexpression of STAT3 in FT secretory epithelial cells promoted tumor progression and metastasis, mimicking the clinical disease observed in patients with HGSC. Thus, we conclude that the STAT3 pathway plays a role in the development and progression of HGSC from its earliest premalignant states.Significance: Concomitant gain of pSTAT3 Tyr705 and loss of PIAS3 appear critical for initiation and development of high-grade serous carcinoma. Cancer Res; 78(7); 1739-50. ©2018 AACR.
Collapse
Affiliation(s)
- Uksha Saini
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Adrian A Suarez
- Department of Pathology, Gynecological Pathology and Cytopathology Unit, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Shan Naidu
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - John J Wallbillich
- Division of Gynecologic Oncology, Department of OB/GYN, Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Kristin Bixel
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Ross A Wanner
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Jason Bice
- Pathology Core Lab, Comprehensive Cancer Center, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Raleigh D Kladney
- Pathology Core Lab, Comprehensive Cancer Center, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Jenny Lester
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California
| | - Beth Y Karlan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars Sinai Medical Center, Los Angeles, California
| | - Paul J Goodfellow
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - David E Cohn
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Karuppaiyah Selvendiran
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, the Ohio State University Wexner Medical Center, Columbus, Ohio.
| |
Collapse
|
22
|
Imran M, Ullah A, Saeed F, Nadeem M, Arshad MU, Suleria HAR. Cucurmin, anticancer, & antitumor perspectives: A comprehensive review. Crit Rev Food Sci Nutr 2017; 58:1271-1293. [PMID: 27874279 DOI: 10.1080/10408398.2016.1252711] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cucurmin, a naturally yellow component isolated from turmeric, ability to prevent various life-style related disorders. The current review article mainly emphasizes on different anticancer perspectives of cucurmin, i.e., colon, cervical, uterine, ovarian, prostate head and neck, breast, pulmonary, stomach and gastric, pancreatic, bladder oral, oesophageal, and bone cancer. It holds a mixture of strong bioactive molecule known as cucurminoids that has ability to reduce cancer/tumor at initial, promotion and progression stages of tumor development. In particular, these compounds block several enzymes required for the growth of tumors and may therefore involve in tumor treatments. Moreover, it modulates an array of cellular progressions, i.e., nitric oxide synthetase activity, protein kinase C activity, epidermal growth factor (EGF) receptor intrinsic kinase activity, nuclear factor kappa (NF-kB) activity, inhibiting lipid peroxidation and production of reactive oxygen species. However, current manuscript summarizes most of the recent investigations of cucurmin but still further research should be conducted to explore the role of curcumin to mitigate various cancers.
Collapse
Affiliation(s)
- Muhammad Imran
- a Department of Diet and Nutritional Sciences , Imperial College of Business Studies , Lahore , Pakistan.,b National Institute of Food Science and Technology , University of Agriculture Faisalabad , Pakistan
| | - Azmat Ullah
- e Department of Food Science and Human Nutrition , University of Veterinary and Animal Sciences , Lahore , Pakistan
| | - Farhan Saeed
- c Institute of Home & Food Sciences , Government College University Faisalabad , Pakistan
| | - Muhammad Nadeem
- d Department of Environmental Sciences , COMSATS Institute of Information Technology Vehari , Pakistan
| | - Muhammad Umair Arshad
- c Institute of Home & Food Sciences , Government College University Faisalabad , Pakistan
| | | |
Collapse
|
23
|
Ko JH, Ho Baek S, Nam D, Chung WS, Lee SG, Lee J, Mo Yang W, Um JY, Seok Ahn K. 3-Formylchromone inhibits proliferation and induces apoptosis of multiple myeloma cells by abrogating STAT3 signaling through the induction of PIAS3. Immunopharmacol Immunotoxicol 2016; 38:334-43. [DOI: 10.1080/08923973.2016.1203928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
24
|
Brassinin inhibits STAT3 signaling pathway through modulation of PIAS-3 and SOCS-3 expression and sensitizes human lung cancer xenograft in nude mice to paclitaxel. Oncotarget 2016; 6:6386-405. [PMID: 25788267 PMCID: PMC4467444 DOI: 10.18632/oncotarget.3443] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/21/2015] [Indexed: 12/14/2022] Open
Abstract
Persistent phosphorylation of signal transducers and activators of transcription 3 (STAT3) is frequently observed in tumor cells. We found that brassinin (BSN) suppressed both constitutive and IL-6-inducible STAT3 activation in lung cancer cells. Moreover, BSN induced PIAS-3 protein and mRNA, whereas the expression of SOCS-3 was reduced. Knockdown of PIAS-3 by small interfering RNA prevented inhibition of STAT3 and cytotoxicity by BSN. Overexpression of SOCS-3 in BSN-treated cells increased STAT3 phosphorylation and cell viability. BSN down-regulated STAT3-regulated gene products, inhibited proliferation, invasion, as well as induced apoptosis. Most importantly, when administered intraperitoneally, combination of BSN and paclitaxel significantly decreased the tumor development in a xenograft lung cancer mouse model associated with down-modulation of phospho-STAT3, Ki-67 and CD31. We suggest that BSN inhibits STAT3 signaling through modulation of PIAS-3 and SOCS-3, thereby attenuating tumor growth and increasing sensitivity to paclitaxel.
Collapse
|
25
|
Yang SF, Hou MF, Chen FM, Ou-Yang F, Wu YC, Chai CY, Yeh YT. Prognostic value of protein inhibitor of activated STAT3 in breast cancer patients receiving hormone therapy. BMC Cancer 2016; 16:20. [PMID: 26768588 PMCID: PMC4714466 DOI: 10.1186/s12885-016-2063-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 01/10/2016] [Indexed: 12/12/2022] Open
Abstract
Background Deregulated signal transducer and activator of transcription 3 (STAT3) signaling has been well documented in certain cancers. Alterations in specific negative regulators, such as protein inhibitor of activated STAT3 (PIAS3), may contribute to cancer development. Methods The expression of total PIAS3 was determined in 100 paired cancerous and non-cancerous breast tissues by immunoblotting and was statistically analyzed along with the clinicopathological characteristics and overall survival of the patients. XTT, immunoblotting, and chromatin immunoprecipitation (Chip) were used to examine the biological effect of PIAS3 in breast cancer cells. Results Hormone therapy failed to improve the overall survival in patients presenting with increased PIAS3 expression. Ectopic PIAS3 overexpression increased the proliferation and expression of cyclin D1 in estrogen receptor (ER)-positive MCF-7 and T47D cells, but decreased those in ER-negative MDA-MB-231 and SKBR3 cells. Furthermore, PIAS3 overexpression attenuated cytotoxicity of tamoxifen and increased proliferation and cyclin D1 expression in MCF-7 cells. PIAS3 also decreased the binding of itself on the cyclin D1 promoter and this decreased binding was not affected by tamoxifen. Conclusion PIAS3 may serve as a biomarker for predicting hormone therapy stratification, although it is limited to those breast cancer patients receiving hormone therapy Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2063-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sheau-Fang Yang
- Department of Pathology, Kaohsiung Municipal Ta-Tung Hospital, No. 68, Zhonghua 3rd Rd, Qianjin Dist, Kaohsiung, 801, R O C, Taiwan.,Department of Pathology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shiquan 1st Rd, Sanmin Dist, Kaohsiung, 807, R O C, Taiwan
| | - Ming-Feng Hou
- Department of Surgery, Kaohsiung Municipal Ta-Tung Hospital, No. 68, Zhonghua 3rd Rd, Qianjin Dist, Kaohsiung, 801, R O C, Taiwan.,Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, No. 100, Shiquan 1st Rd, Sanmin Dist, Kaohsiung, 807, R O C, Taiwan
| | - Fang-Ming Chen
- Department of Surgery, Kaohsiung Municipal Ta-Tung Hospital, No. 68, Zhonghua 3rd Rd, Qianjin Dist, Kaohsiung, 801, R O C, Taiwan.,Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, No. 100, Shiquan 1st Rd, Sanmin Dist, Kaohsiung, 807, R O C, Taiwan.,Department of Laboratory, Kaohsiung Municipal Ta-Tung Hospital, No. 68, Zhonghua 3rd Rd, Qianjin Dist, Kaohsiung, 801, R O C, Taiwan
| | - Fu Ou-Yang
- Department of Surgery, Kaohsiung Municipal Ta-Tung Hospital, No. 68, Zhonghua 3rd Rd, Qianjin Dist, Kaohsiung, 801, R O C, Taiwan.,Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, No. 100, Shiquan 1st Rd, Sanmin Dist, Kaohsiung, 807, R O C, Taiwan
| | - Yang-Chang Wu
- School of Pharmacy, College of Pharmacy, China Medical University, No. 91, Hsueh-Shih Road, Taichung, R O C, 40402, Taiwan.,Chinese Medicine Research and Development Center, China Medical University Hospital, No. 2, Yude Road, Taichung, 40447, R O C, Taiwan
| | - Chee-Yin Chai
- Department of Pathology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shiquan 1st Rd, Sanmin Dist, Kaohsiung, 807, R O C, Taiwan.
| | - Yao-Tsung Yeh
- Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, No.151, Jinxue Rd, Daliao Dist, Kaohsiung, 831, R O C, Taiwan.
| |
Collapse
|
26
|
Lai PS, Rosa DA, Magdy Ali A, Gómez-Biagi RF, Ball DP, Shouksmith AE, Gunning PT. A STAT inhibitor patent review: progress since 2011. Expert Opin Ther Pat 2015; 25:1397-421. [PMID: 26394986 DOI: 10.1517/13543776.2015.1086749] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The clinical utility of effective direct STAT inhibitors, particularly STAT3 and STAT5, for treating cancer and other diseases is well studied and known. AREAS COVERED This review will highlight the STAT inhibitor patent literature from 2011 to 2015 inclusive. Emphasis will be placed on inhibitors of the STAT3, STAT5a/b, and STAT1 proteins for cancer treatment. The review will, where suitably investigated, describe the mode and the site of inhibition, list indications that were evaluated, and rank the inhibitor's relative potency among compounds in the same class. The reader will gain an understanding of the diverse set of approaches, used both in academia and industry, to target STAT proteins. EXPERT OPINION There is still much work to be done to directly target the STAT3 and STAT5 proteins. As yet, there is still no direct STAT3 inhibitor in the clinic. While the SH2 domain remains a popular target for therapeutic intervention, the DNA-binding domain and N-terminal region are now attracting attention as possible sites for inhibition. Multiple putative STAT3 and STAT5 inhibitors have now been patented across a broad spectrum of chemotypes, each with their own advantages and limitations.
Collapse
Affiliation(s)
- Ping-Shan Lai
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| | - David A Rosa
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| | - Ahmed Magdy Ali
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| | - Rodolfo F Gómez-Biagi
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| | - Daniel P Ball
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| | - Andrew E Shouksmith
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| | - Patrick T Gunning
- a University of Toronto Mississauga, Department of Chemical and Physical Sciences , 3359 Mississauga Road North, Mississauga, Ontario L5L 1C6, Canada +1 90 55 69 45 88 ; +1 90 55 69 49 29 ;
| |
Collapse
|
27
|
Schütz A, Röser K, Klitzsch J, Lieder F, Aberger F, Gruber W, Mueller KM, Pupyshev A, Moriggl R, Friedrich K. Lung Adenocarcinomas and Lung Cancer Cell Lines Show Association of MMP-1 Expression With STAT3 Activation. Transl Oncol 2015; 8:97-105. [PMID: 25926075 PMCID: PMC4415137 DOI: 10.1016/j.tranon.2015.02.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 01/21/2015] [Accepted: 02/04/2015] [Indexed: 01/10/2023] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is constitutively activated in the majority of lung cancer. This study aims at defining connections between STAT3 function and the malignant properties of non–small cell lung carcinoma (NSCLC) cells. To address possible mechanisms by which STAT3 influences invasiveness, the expression of matrix metalloproteinase-1 (MMP-1) was analyzed and correlated with the STAT3 activity status. Studies on both surgical biopsies and on lung cancer cell lines revealed a coincidence of STAT3 activation and strong expression of MMP-1. MMP-1 and tyrosine-phosphorylated activated STAT3 were found co-localized in cancer tissues, most pronounced in tumor fronts, and in particular in adenocarcinomas. STAT3 activity was constitutive, although to different degrees, in the lung cancer cell lines investigated. Three cell lines (BEN, KNS62, and A549) were identified in which STAT3 activitation was inducible by Interleukin-6 (IL-6). In A549 cells, STAT3 activity enhanced the level of MMP-1 mRNA and stimulated transcription from the MMP-1 promoter in IL-6–stimulated A549 cells. STAT3 specificity of this effect was confirmed by STAT3 knockdown through RNA interference. Our results link aberrant activity of STAT3 in lung cancer cells to malignant tumor progression through up-regulation of expression of invasiveness-associated MMPs.
Collapse
Affiliation(s)
| | - Katrin Röser
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany
| | - Jana Klitzsch
- Institute of Pathology, University of Leipzig, Leipzig, Germany
| | - Franziska Lieder
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany
| | - Fritz Aberger
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Wolfgang Gruber
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | | | | | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | | |
Collapse
|
28
|
Pastuszak-Lewandoska D, Domańska D, Czarnecka KH, Kordiak J, Migdalska-Sęk M, Nawrot E, Kiszałkiewicz J, Antczak A, Górski P, Brzeziańska E. Expression of STAT5, COX-2 and PIAS3 in correlation with NSCLC histhopathological features. PLoS One 2014; 9:e104265. [PMID: 25137041 PMCID: PMC4138089 DOI: 10.1371/journal.pone.0104265] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 07/11/2014] [Indexed: 11/22/2022] Open
Abstract
Signal transducers and activators of transcription (STATs), their inhibitors and cyclooxygenase-2 (COX-2) participate in transformations of many various types of cancers. The aim of the present study was to evaluate the relationship between STAT5A/B, COX-2, and PIAS3 mRNA expression and tumor staging, metastasis status, and histopathological subtype in 71 patients with confirmed non-small cell lung cancer (NSCLC) diagnosis. Total RNA was isolated from NSCLC tissue samples and the expression of the studied genes was assessed using TaqMan probes in real-time PCR assay. The expression levels of STAT5A, STAT5B, and COX-2 genes were increased in 69%, 79%, and 71% NSCLC samples respectively, while PIAS3 expression was decreased in the majority (69%) of the studied tissues. Statistically significant differences were observed between STAT5 isoforms (P = 0.0008), with higher expression of STAT5B. We found statistically significant positive correlation between STAT5B and COX-2 (rho = 0.045), and significant negative correlation between STAT5B and PIAS3 (rho = −0.049). The negative correlation between STAT5B and PIAS3 (rho = −0.43) was also observed in T2a+T2b tumor group. Additionally, STAT5B and COX-2 expression levels were significantly different between T1a+T1b and T2a+T2b tumors (P = 0.002 and P = 0.041, respectively), with higher expression of both genes in T2 tumor stage. PIAS3 expression was significantly lower in NSCC subtype as compared with SCC subtype (P = 0.017). Also, STAT5A and STAT5B immunoexpression was assessed, and the results indicated significantly higher protein levels in NSCLC patients as compared with controls (P = 0.048 and P = 0.034, respectively). High STAT5B immunoexpression was positively correlated with STAT5B gene expression in tumors (rho = 0.755). STAT5B protein level was also significantly higher in T2a+T2b tumors, reflecting high STAT5B gene expression in this group. There was no statistically significant association between mRNA and protein expression levels of the studied genes and patients' characteristics: age, gender, smoking. The obtained results highlight the importance of the genes STAT5B and COX-2 in lung cancer progression.
Collapse
Affiliation(s)
| | - Daria Domańska
- Department of Molecular Bases of Medicine, Medical University of Lodz, Lodz, Poland
| | | | - Jacek Kordiak
- Department of Chest Surgery, General and Oncological Surgery, University Hospital No. 2, Medical University of Lodz, Lodz, Poland
| | - Monika Migdalska-Sęk
- Department of Molecular Bases of Medicine, Medical University of Lodz, Lodz, Poland
| | - Ewa Nawrot
- Department of Molecular Bases of Medicine, Medical University of Lodz, Lodz, Poland
| | | | - Adam Antczak
- Department of General and Oncological Pulmonology, Medical University of Lodz, Lodz, Poland
| | - Paweł Górski
- Department of Pneumology and Allergology, Medical University of Lodz, Lodz, Poland
| | - Ewa Brzeziańska
- Department of Molecular Bases of Medicine, Medical University of Lodz, Lodz, Poland
- * E-mail:
| |
Collapse
|
29
|
Dabir S, Kluge A, Kresak A, Yang M, Fu P, Groner B, Wildey G, Dowlati A. Low PIAS3 expression in malignant mesothelioma is associated with increased STAT3 activation and poor patient survival. Clin Cancer Res 2014; 20:5124-32. [PMID: 25124686 DOI: 10.1158/1078-0432.ccr-14-1233] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Deregulation of STAT3 activation is a hallmark of many cancer cells, and the underlying mechanisms are subject to intense investigation. We examined the extent of PIAS3 expression in mesothelioma cells and human tumor samples and determined the functional effects of PIAS3 expression on STAT3 signaling. EXPERIMENTAL DESIGN We evaluated the expression of PIAS3 in mesothelioma tumors from patients and correlated the expression levels with the course of the disease. We also measured the effects of enhanced PIAS3 activity on STAT3 signaling, cellular growth, and viability in cultured mesothelioma cells. RESULTS Gene expression databases revealed that mesotheliomas have the lowest levels of PIAS3 transcripts among solid tumors. PIAS3 expression in human mesothelioma tumors is significantly correlated with overall survival intervals (P = 0.058). The high expression of PIAS3 is predictive of a favorable prognosis and decreases the probability of death within one year after diagnosis by 44%. PIAS3 expression is functionally linked to STAT3 activation in mesothelioma cell lines. STAT3 downregulation with siRNA or enhanced expression of PIAS3 both inhibited mesothelioma cell growth and induced apoptosis. Mesothelioma cells are sensitive to curcumin and respond by the induction of PIAS3. Corroborative evidence has been obtained from STAT3 inhibition experiments. Exposure of the cells to a peptide derived from the PIAS3 protein that interferes with STAT3 function resulted in apoptosis induction and the inhibition of cell growth. CONCLUSION These results suggest that PIAS3 protein expression impacts survival in patients with mesothelioma and that PIAS3 activation could become a therapeutic strategy. Clin Cancer Res; 20(19); 5124-32. ©2014 AACR.
Collapse
Affiliation(s)
- Snehal Dabir
- Division of Hematology and Oncology, Case Comprehensive Cancer Center and University Hospitals Case Medical Center, Cleveland, Ohio
| | - Amy Kluge
- Division of Hematology and Oncology, Case Comprehensive Cancer Center and University Hospitals Case Medical Center, Cleveland, Ohio
| | - Adam Kresak
- Division of Pathology, Case Comprehensive Cancer Center and University Hospitals Case Medical Center, Cleveland, Ohio
| | - Michael Yang
- Division of Pathology, Case Comprehensive Cancer Center and University Hospitals Case Medical Center, Cleveland, Ohio
| | - Pingfu Fu
- Division of Biostatistics, Case Comprehensive Cancer Center and University Hospitals Case Medical Center, Cleveland, Ohio
| | - Bernd Groner
- Institute for Biomedical Research, Georg-Speyer-Haus, Frankfurt, Germany
| | - Gary Wildey
- Division of Hematology and Oncology, Case Comprehensive Cancer Center and University Hospitals Case Medical Center, Cleveland, Ohio
| | - Afshin Dowlati
- Division of Hematology and Oncology, Case Comprehensive Cancer Center and University Hospitals Case Medical Center, Cleveland, Ohio.
| |
Collapse
|
30
|
STAT3 in Cancer-Friend or Foe? Cancers (Basel) 2014; 6:1408-40. [PMID: 24995504 PMCID: PMC4190548 DOI: 10.3390/cancers6031408] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 06/19/2014] [Accepted: 06/20/2014] [Indexed: 12/25/2022] Open
Abstract
The roles and significance of STAT3 in cancer biology have been extensively studied for more than a decade. Mounting evidence has shown that constitutive activation of STAT3 is a frequent biochemical aberrancy in cancer cells, and this abnormality directly contributes to tumorigenesis and shapes many malignant phenotypes in cancer cells. Nevertheless, results from more recent experimental and clinicopathologic studies have suggested that STAT3 also can exert tumor suppressor effects under specific conditions. Importantly, some of these studies have demonstrated that STAT3 can function either as an oncoprotein or a tumor suppressor in the same cell type, depending on the specific genetic background or presence/absence of specific coexisting biochemical defects. Thus, in the context of cancer biology, STAT3 can be a friend or foe. In the first half of this review, we will highlight the “evil” features of STAT3 by summarizing its oncogenic functions and mechanisms. The differences between the canonical and non-canonical pathway will be highlighted. In the second half, we will summarize the evidence supporting that STAT3 can function as a tumor suppressor. To explain how STAT3 may mediate its tumor suppressor effects, we will discuss several possible mechanisms, one of which is linked to the role of STAT3β, one of the two STAT3 splicing isoforms. Taken together, it is clear that the roles of STAT3 in cancer are multi-faceted and far more complicated than one appreciated previously. The new knowledge has provided us with new approaches and strategies when we evaluate STAT3 as a prognostic biomarker or therapeutic target.
Collapse
|
31
|
Yu T, Lu Q, Ou X, Cao D, Yu Q. Association of sedentary behavior with the expression levels of biomarkers in colorectal cancer: clinical analysis of 228 patients. TOHOKU J EXP MED 2014; 232:167-76. [PMID: 24621823 DOI: 10.1620/tjem.232.167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
There is an association between sedentary behavior and the risk of colorectal cancer (CRC), but the underlying mechanism is unclear. CRC is characterized by the changes in the expression levels of biomarkers, including voltage-gated proton channel Hv1, matrix-remodeling associated 5 (MXRA5), DEK (inducing positive supercoils into circular DNA) and protein inhibitor of activated signal transducer and activators of transcription 3 (PIAS3). Thus, sedentary behavior may affect the expression levels of these biomarkers in the colorectal tissue. Here, we recruited 228 CRC patients (128 males, 57.8 ± 7.8 years; 100 females, 57.7 ± 7.5 years) and 80 healthy subjects (48 males, 57.5 ± 6.8 years; 32 females, 56.9 ± 6.5 years) from March 7th, 2010 to May 6th, 2012. All the subjects were unrelated Han Chinese with the similar cultural and economic background. All the subjects were interviewed concerning sedentary time (sitting time categories: less than 1, 1-3, 4-6, and more than 6 h/day). The daily sedentary time of most CRC patients was more than 4 h/day, while the sedentary time of most healthy subjects was less than 3 h/day. The expression levels of Hv1, MXRA5 and DEK mRNAs and proteins were higher in CRC tissues and the levels of PIAS3 mRNA and protein were lower when the daily sedentary time was longer in CRC patients (p < 0.05). The daily sedentary time was correlated with the protein levels of CRC biomarkers. Furthermore, the sedentary time was positively related with body mass index but not daily calorie intake.
Collapse
Affiliation(s)
- Ting Yu
- Department of Gastroenterology, Affiliated Zhongda Hospital of Southeast University
| | | | | | | | | |
Collapse
|
32
|
Peng Q, Schork NJ. Utility of network integrity methods in therapeutic target identification. Front Genet 2014; 5:12. [PMID: 24550933 PMCID: PMC3909879 DOI: 10.3389/fgene.2014.00012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 01/13/2014] [Indexed: 01/05/2023] Open
Abstract
Analysis of the biological gene networks involved in a disease may lead to the identification of therapeutic targets. Such analysis requires exploring network properties, in particular the importance of individual network nodes (i.e., genes). There are many measures that consider the importance of nodes in a network and some may shed light on the biological significance and potential optimality of a gene or set of genes as therapeutic targets. This has been shown to be the case in cancer therapy. A dilemma exists, however, in finding the best therapeutic targets based on network analysis since the optimal targets should be nodes that are highly influential in, but not toxic to, the functioning of the entire network. In addition, cancer therapeutics targeting a single gene often result in relapse since compensatory, feedback and redundancy loops in the network may offset the activity associated with the targeted gene. Thus, multiple genes reflecting parallel functional cascades in a network should be targeted simultaneously, but require the identification of such targets. We propose a methodology that exploits centrality statistics characterizing the importance of nodes within a gene network that is constructed from the gene expression patterns in that network. We consider centrality measures based on both graph theory and spectral graph theory. We also consider the origins of a network topology, and show how different available representations yield different node importance results. We apply our techniques to tumor gene expression data and suggest that the identification of optimal therapeutic targets involving particular genes, pathways and sub-networks based on an analysis of the nodes in that network is possible and can facilitate individualized cancer treatments. The proposed methods also have the potential to identify candidate cancer therapeutic targets that are not thought to be oncogenes but nonetheless play important roles in the functioning of a cancer-related network or pathway.
Collapse
Affiliation(s)
- Qian Peng
- Department of Molecular and Experimental Medicine, The Scripps Research Institute La Jolla, CA, USA ; Scripps Genomic Medicine, The Scripps Translational Science Institute La Jolla, CA, USA
| | - Nicholas J Schork
- Department of Molecular and Experimental Medicine, The Scripps Research Institute La Jolla, CA, USA ; Scripps Genomic Medicine, The Scripps Translational Science Institute La Jolla, CA, USA
| |
Collapse
|
33
|
Li H, Gao H, Bijukchhe SM, Wang Y, Li T. PIAS3 may represent a potential biomarker for diagnosis and therapeutic of human colorectal cancer. Med Hypotheses 2013; 81:1151-4. [PMID: 24120699 DOI: 10.1016/j.mehy.2013.09.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 08/30/2013] [Accepted: 09/15/2013] [Indexed: 01/22/2023]
Abstract
Colorectal cancer (CRC) is a challenging problem both for the developed and underdeveloped countries. Despite numerous improvements in early diagnosis and treatment, the incidence and mortality is still keeping in a high level. Molecule targeted therapy has drawn much attention as next generation anticancer agents for diagnosis and therapeutic of CRC. Protein Inhibitor of Activated Signal Transducer and Activators of Transcription 3 (PIAS3) as a novel biomarker has been focused to have a role in the development of malignancy, which was expressed at a higher level in most common malignancies compared with corresponding normal tissues. Furthermore, evidences suggest that the expression of PIAS3 can affect the growth of cancer cells by inhibiting the JAK/STAT and PI3-K/Akt signaling pathways or regulating its SUMO (small-ubiquitin like modifiers) ligase activity in some malignancy. Therefore, we hypothesized that PIAS3 may be a potential biomarker target for early cancer detection and therapeutic of human CRC.
Collapse
Affiliation(s)
- Heping Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital to Xinjiang Medical University, Urumqi 830054, China
| | | | | | | | | |
Collapse
|
34
|
Mautsa N, Prinsloo E, Bishop ÖT, Blatch GL. The PINIT domain of PIAS3: structure-function analysis of its interaction with STAT3. J Mol Recognit 2012; 24:795-803. [PMID: 21812053 DOI: 10.1002/jmr.1111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The protein inhibitor of activated signal transducer and activator of transcription 3 (PIAS3) regulates the transcriptional activity of signal transducer and activator of transcription 3 (STAT3) which regulates transcription of genes involved in cell growth, proliferation and apoptosis. The conserved proline, isoleucine, asparagine, isoleucine, threonine (PINIT) domain of PIAS3 is thought to promote STAT3-PIAS3 interaction. The (His)(7) -PINIT domain (PIAS3(85-272) ) was heterologously expressed and purified to homogeneity by nickel affinity and size exclusion chromatography, and shown to be a folded monomer in solution. Using surface plasmon resonance spectroscopy (SPR) the PINIT domain (PIAS3(85-272) ) alone was shown to specifically bind to STAT3 in a concentration dependent manner. L97A, R99N and R99Q mutations of the PINIT domain were found to abrogate binding to STAT3, suggesting that these residues were part of a potential binding surface. An homology model for the PINIT domain was calculated to analyse the potential locations of L97 and R99 in the structure, and to evaluate the potential role of these residues in interactions with STAT3.
Collapse
Affiliation(s)
- Nicodemus Mautsa
- Biomedical Biotechnology Research Unit, Department of Biochemistry, Microbiology and Biotechnology, Rhodes University, Grahamstown 6140, South Africa
| | | | | | | |
Collapse
|
35
|
Shu XH, Li H, Sun XX, Wang Q, Sun Z, Wu ML, Chen XY, Li C, Kong QY, Liu J. Metabolic patterns and biotransformation activities of resveratrol in human glioblastoma cells: relevance with therapeutic efficacies. PLoS One 2011; 6:e27484. [PMID: 22096581 PMCID: PMC3214056 DOI: 10.1371/journal.pone.0027484] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 10/17/2011] [Indexed: 01/19/2023] Open
Abstract
Background Trans-resveratrol rather than its biotransformed monosulfate metabolite exerts anti-medulloblastoma effects by suppressing STAT3 activation. Nevertheless, its effects on human glioblastoma cells are variable due to certain unknown reason(s). Methodology/Principal Findings Citing resveratrol-sensitive UW228-3 medulloblastoma cell line and primarily cultured rat brain cells/PBCs as controls, the effect of resveratrol on LN-18 human glioblastoma cells and its relevance with metabolic pattern(s), brain-associated sulfotransferase/SULT expression and the statuses of STAT3 signaling and protein inhibitor of activated STAT3 (PIAS3) were elucidated by multiple experimental approaches. Meanwhile, the expression patterns of three SULTs (SULT1A1, 1C2 and 4A1) in human glioblastoma tumors were profiled immunohistochemically. The results revealed that 100 µM resveratrol-treated LN-18 generated the same metabolites as UW228-3 cells, while additional metabolite in molecular weight of 403.0992 in negative ion mode was found in PBCs. Neither growth arrest nor apoptosis was found in resveratrol-treated LN-18 and PBC cells. Upon resveratrol treatment, the levels of SULT1A1, 1C2 and 4A1 expression in LN-18 cells were more up-regulated than that expressed in UW228-3 cells and close to the levels in PBCs. Immunohistochemical staining showed that 42.0%, 27.1% and 19.6% of 149 glioblastoma cases produced similar SULT1A1, 1C2 and 4A1 levels as that of tumor-surrounding tissues. Unlike the situation in UW228-3 cells, STAT3 signaling remained activated and its protein inhibitor PIAS3 was restricted in the cytosol of resveratrol-treated LN-18 cells. No nuclear translocation of STAT3 and PIAS3 was observed in resveratrol-treated PBCs. Treatment with STAT3 chemical inhibitor, AG490, committed majority of LN-18 and UW228-3 cells but not PBCs to apoptosis within 48 hours. Conclusions/Significance LN-18 glioblastoma cells are insensitive to resveratrol due to the more inducible brain-associated SULT expression, insufficiency of resveratrol to suppress activated STAT3 signaling and the lack of PIAS3 nuclear translocation. The findings from PBCs suggest that an effective anticancer dose of resveratrol exerts little side effect on normal brain cells.
Collapse
Affiliation(s)
- Xiao-Hong Shu
- Liaoning Laboratory of Cancer Genetics & Epigenetics, Department of Cell Biology, Dalian Medical University, Dalian, China
- Department of Medicinal Chemistry, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Hong Li
- Liaoning Laboratory of Cancer Genetics & Epigenetics, Department of Cell Biology, Dalian Medical University, Dalian, China
| | - Xiao-Xin Sun
- Liaoning Laboratory of Cancer Genetics & Epigenetics, Department of Cell Biology, Dalian Medical University, Dalian, China
| | - Qian Wang
- Liaoning Laboratory of Cancer Genetics & Epigenetics, Department of Cell Biology, Dalian Medical University, Dalian, China
| | - Zheng Sun
- Liaoning Laboratory of Cancer Genetics & Epigenetics, Department of Cell Biology, Dalian Medical University, Dalian, China
| | - Mo-Li Wu
- Liaoning Laboratory of Cancer Genetics & Epigenetics, Department of Cell Biology, Dalian Medical University, Dalian, China
| | - Xiao-Yan Chen
- Liaoning Laboratory of Cancer Genetics & Epigenetics, Department of Cell Biology, Dalian Medical University, Dalian, China
| | - Chong Li
- Liaoning Laboratory of Cancer Genetics & Epigenetics, Department of Cell Biology, Dalian Medical University, Dalian, China
| | - Qing-You Kong
- Liaoning Laboratory of Cancer Genetics & Epigenetics, Department of Cell Biology, Dalian Medical University, Dalian, China
| | - Jia Liu
- Liaoning Laboratory of Cancer Genetics & Epigenetics, Department of Cell Biology, Dalian Medical University, Dalian, China
- * E-mail:
| |
Collapse
|
36
|
Liu LM, Yan MG, Yang DH, Sun WW, Zhang JX. The expression of protein inhibitor of activated signal transducers and activators of transcription 3 in the evolutionary process of gastric cancer. Eur J Intern Med 2011; 22:e31-e35. [PMID: 21925039 DOI: 10.1016/j.ejim.2011.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 04/03/2011] [Accepted: 04/04/2011] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To study the expression of PIAS3 (protein inhibitor of activated signal transducers and activators of transcription 3) in the evolutionary process of gastric cancer. METHODS Samples were taken from the endoscopic biopsy specimens of 125 patients. Gastric mucosal lesions were diagnosed in HE staining, and chronic atrophic gastritis (CAG) with intestinal metaplasia (IM) were distinguished in AB-PAS and HID-AB staining. The expressions of PIAS3 gene in different types of gastric mucosal lesions were detected by immunocytochemistry and in situ hybridization. The results were analyzed using IPP 6.0 image analysis system, from which the average optical density was obtained of positive cells. RESULTS There were 25 patients with chronic superficial gastritis (CSG), 87 CAG (30 with complete intestinal IM, 27 with incomplete intestinal IM, 21 with complete colonic IM, 9 with incomplete colonic IM), 8 dysplasia (DYS) and 5 gastric cancer (GC). In the expressions of PIAS3 mRNA and protein, a difference was not found between the patients with CSG and those with CAG with complete or incomplete intestinal IM; however, a significant difference was statistically found among patients with CSG (or intestinal IM), complete colonic IM, incomplete colonic IM, DYS and GC, expression levels of which stepped down one by one. CONCLUSIONS There are differences in the PIAS3 expression from different stages of gastric precancerous conditions/lesions to GC, which may reveal a close relationship between expression reduction or loss of PIAS3 and gastric tumorigenesis.
Collapse
Affiliation(s)
- Liang-Ming Liu
- Songjiang Hospital Affiliated to the First People's Hospital Shanghai Jiaotong University, Shanghai 201600, China.
| | | | | | | | | |
Collapse
|
37
|
Suda N, Shibata H, Kurihara I, Ikeda Y, Kobayashi S, Yokota K, Murai-Takeda A, Nakagawa K, Oya M, Murai M, Rainey WE, Saruta T, Itoh H. Coactivation of SF-1-mediated transcription of steroidogenic enzymes by Ubc9 and PIAS1. Endocrinology 2011; 152:2266-77. [PMID: 21467194 PMCID: PMC3100613 DOI: 10.1210/en.2010-1232] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 03/10/2011] [Indexed: 01/07/2023]
Abstract
Steroidogenic factor-1 (SF-1) is a nuclear orphan receptor, which is essential for adrenal development and regulation of steroidogenic enzyme expression. SF-1 is posttranslationally modified by small ubiquitin-related modifier-1 (SUMO-1), thus mostly resulting in attenuation of transcription. We investigated the role of sumoylation enzymes, Ubc9 and protein inhibitors of activated STAT1 (PIAS1), in SF-1-mediated transcription of steroidogenic enzyme genes in the adrenal cortex. Coimmunoprecipitation assays showed that both Ubc9 and PIAS1 interacted with SF-1. Transient transfection assays in adrenocortical H295R cells showed Ubc9 and PIAS1 potentiated SF-1-mediated transactivation of reporter constructs containing human CYP17, CYP11A1, and CYP11B1 but not CYP11B2 promoters. Reduction of endogenous Ubc9 and PIAS1 by introducing corresponding small interfering RNA significantly reduced endogenous CYP17, CYP11A1, and CYP11B1 mRNA levels, indicating that they normally function as coactivators of SF-1. Wild type and sumoylation-inactive mutants of Ubc9 and PIAS1 can similarly enhance the SF-1-mediated transactivation of the CYP17 gene, indicating that the coactivation potency of Ubc9 and PIAS1 is independent of sumoylation activity. Chromatin immunoprecipitation assays demonstrated that SF-1, Ubc9, and PIAS1 were recruited to an endogenous CYP17 gene promoter in the context of chromatin in vivo. Immunohistochemistry and Western blotting showed that SF-1, Ubc9, and PIAS1 were expressed in the nuclei of the human adrenal cortex. In cortisol-producing adenomas, the expression pattern of SF-1 and Ubc9 were markedly increased, whereas that of PIAS1 was decreased compared with adjacent normal adrenals. These results showed the physiological roles of Ubc9 and PIAS1 as SF-1 coactivators beyond sumoylation enzymes in adrenocortical steroidogenesis and suggested their possible pathophysiological roles in human cortisol-producing adenomas.
Collapse
Affiliation(s)
- Noriko Suda
- Department of Internal Medicine, School of Medicine, Keio University, Shinjujku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kluge A, Dabir S, Vlassenbroeck I, Eisenberg R, Dowlati A. Protein inhibitor of activated STAT3 expression in lung cancer. Mol Oncol 2011; 5:256-64. [PMID: 21497567 PMCID: PMC3104085 DOI: 10.1016/j.molonc.2011.03.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 03/17/2011] [Accepted: 03/17/2011] [Indexed: 01/22/2023] Open
Abstract
Protein Inhibitor of Activated Signal Transducer and Activators of Transcription 3 (PIAS3) is an endogenous inhibitor of STAT3 transcriptional activity. We have previously demonstrated the concentration-dependent negative regulatory effect of PIAS3 on STAT3 signaling and its capacity to decrease lung cancer proliferation and synergize with epidermal growth factor inhibition. We now investigate PIAS3 expression in both non-small cell lung cancer (NSCLC) cell lines and human resected NSCLC specimens. We also investigated the mechanism by which some lung cancers have significantly decreased PIAS3 expression. Expression of PIAS3 is variable in lung cancer cells lines with 2 of 3 squamous cell carcinoma (SCC) cell lines having no or little PIAS3 protein expression. Similarly, the majority of human SCCs of the lung lack PIAS3 expression by immunohistochemistry; this despite the finding that SCCs have significantly higher levels of PIAS3 mRNA compared to adenocarcinomas. High PIAS3 expression generally correlates with decreased phosphorylated STAT3 in both SCC cell lines and human specimens compatible with the negative regulatory effect of this protein on STAT3 signaling. To investigate this variable expression of PIAS3 we first performed sequencing of the PIAS3 gene that demonstrated single nucleotide polymorphisms but no mutations. Exposure of lung cancer cells to 5-azacytidine and trichostatin A results in a significant increase in PIAS3 mRNA and protein expression. However, methylation-specific PCR demonstrates a lack of CpG island methylation in the promoter region of PIAS3. Exposure of cells to an agent blocking proteosomal degradation results in a significant increase in PIAS3. Our data thus shows that SCC of the lung commonly lacks PIAS3 protein expression and that post-translational modifications may explain this finding in some cases. PIAS3 is a potential therapeutic molecule to target STAT3 pathway in lung cancer.
Collapse
MESH Headings
- Acetylation
- Base Sequence
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/surgery
- Carcinoma, Squamous Cell/genetics
- Cell Line, Tumor
- DNA Methylation/genetics
- Gene Expression Regulation, Neoplastic
- Histones/metabolism
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Lung Neoplasms/surgery
- Molecular Chaperones/genetics
- Molecular Chaperones/metabolism
- Molecular Sequence Data
- Mutation/genetics
- Neoplasm Staging
- Polymorphism, Single Nucleotide/genetics
- Promoter Regions, Genetic/genetics
- Proteasome Endopeptidase Complex/metabolism
- Protein Inhibitors of Activated STAT/genetics
- Protein Inhibitors of Activated STAT/metabolism
- Protein Processing, Post-Translational
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Sequence Analysis, DNA
Collapse
Affiliation(s)
- Amy Kluge
- Division of Hematology/Oncology (AK, SD, AD), Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States
| | - Snehal Dabir
- Division of Hematology/Oncology (AK, SD, AD), Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States
| | | | - Rosana Eisenberg
- Department of Pathology (RE), Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States
| | - Afshin Dowlati
- Division of Hematology/Oncology (AK, SD, AD), Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, United States
| |
Collapse
|
39
|
Liu LM, Yan MG, Yang DH, Sun WW, Zhang JX. PIAS3 expression in human gastric carcinoma and its adjacent non-tumor tissues. Clin Res Hepatol Gastroenterol 2011; 35:393-398. [PMID: 21334998 DOI: 10.1016/j.clinre.2010.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 11/27/2010] [Accepted: 12/02/2010] [Indexed: 02/04/2023]
Abstract
OBJECTIVE PIAS3 is the endogenous inhibitor of STAT3, which has been implicated in the pathogenesis of many cancers. However, the effect of PIAS3 on human tumors remains elusive. The aim of this article is to investigate the expression of PIAS3 in gastric carcinoma and its adjacent non-tumor tissues. METHODS Samples were taken from 30 patients with gastric cancer, which included tumor or non-tumor tissues in the excised sections. The expression of PIAS3 protein was detected by immunocytochemistry, and that of mRNA by in situ hybridization. The results were semi-quantitative analyzed by using cell count and color depth to stage. RESULTS The expression levels of PIAS3 protein and mRNA were significantly lower in gastric cancerous tissues than in its adjacent non-tumor tissues, and had a close relation with tumor size and differentiation, but not with age, gender and lymphatic metastasis in gastric carcinoma. The more large in size and poorly in differentiation, the more low PIAS3 expression was. CONCLUSION Loss of PIAS3 expression may be an important characteristic of gastric cancer and suggest vicious degree of the tumor.
Collapse
Affiliation(s)
- Liang-ming Liu
- Songjiang Hospital Affiliated to The First People's Hospital Shanghai Jiaotong University, Shanghai 201600, China.
| | | | | | | | | |
Collapse
|
40
|
Saydmohammed M, Joseph D, Syed V. Curcumin suppresses constitutive activation of STAT-3 by up-regulating protein inhibitor of activated STAT-3 (PIAS-3) in ovarian and endometrial cancer cells. J Cell Biochem 2010; 110:447-56. [PMID: 20235152 DOI: 10.1002/jcb.22558] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Signal transducer and activator of transcription-3 (STAT-3) is constitutively activated in ovarian and endometrial cancers and is implicated in uncontrolled cell growth. Thus, its disruption could be an effective approach to control tumorigenesis. Curcumin is a dihydroxyphenolic compound, with proven anti-cancer efficacy in various cancer models. We examined the anti-tumor mechanism of curcumin on STAT-3 and on the negative regulators of STAT-3, including suppressors of cytokine signaling proteins (SOCS-1 and SOCS-3), protein inhibitors of activated STAT (PIAS-1 and PIAS-3), and SH2 domain-containing phosphatases (SHP-1 and SHP-2) in ovarian and endometrial cancer cell lines. Treatment of cancer cells with curcumin induced a dose- and time-dependent decrease of constitutive IL-6 expression and of constitutive and IL-6-induced STAT-3 phosphorylation, which is associated with decreased cell viability and increased cleavage of caspase-3. The inhibition of STAT-3 activation by curcumin was reversible, and phosphorylated STAT-3 levels returned to control levels 24 h after curcumin removal. Compared to normal cells baseline expression of SOCS-3 was high in cancer cells and a marked decrease in SOCS-3 expression was seen following curcumin treatment. Overexpression of SOCS-3 in curcumin-treated cells increased expression of phosphorylated STAT-3 and resulted in increased cell viability. Normal ovarian and endometrial cells exhibited high expression of PIAS-3 protein, whereas in cancer cells the expression was greatly reduced. Curcumin increased PIAS-3 expression in cancer cells. Of significance, siRNA-mediated knockdown of PIAS-3 overcomes the inhibitory effect of curcumin on STAT-3 phosphorylation and cell viability. In conclusion, curcumin suppresses JAK-STAT signaling via activation of PIAS-3, thus attenuating STAT-3 phosphorylation and tumor cell growth.
Collapse
Affiliation(s)
- Manush Saydmohammed
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814-4799, USA
| | | | | |
Collapse
|
41
|
Lai SY, Johnson FM. Defining the role of the JAK-STAT pathway in head and neck and thoracic malignancies: implications for future therapeutic approaches. Drug Resist Updat 2010; 13:67-78. [PMID: 20471303 DOI: 10.1016/j.drup.2010.04.001] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Accepted: 04/06/2010] [Indexed: 12/17/2022]
Abstract
Although the role of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway has been most extensively studied in hematopoietic cells and hematologic malignancies, it is also activated in epithelial tumors, including those originating in the lungs and head and neck. The canonical pathway involves the activation of JAK following ligand binding to cytokine receptors. The activated JAKs then phosphorylate STAT proteins, leading to their dimerization and translocation into the nucleus. In the nucleus, STATs act as transcription factors with pleiotropic downstream effects. STATs can be activated independently of JAKs, most notably by c-Src kinases. In cancer cells, STAT3 and STAT5 activation leads to the increased expression of downstream target genes, leading to increased cell proliferation, cell survival, angiogenesis, and immune system evasion. STAT3 and STAT5 are expressed and activated in head and neck squamous cell carcinoma (HNSCC) where they contribute to cell survival and proliferation. In HNSCC, STATs can be activated by a number of signal transduction pathways, including the epidermal growth factor receptor (EGFR), alpha7 nicotinic receptor, interleukin (IL) receptor, and erythropoietin receptor pathways. Activated STATs are also expressed in lung cancer, but the biological effects of JAK/STAT inhibition in this cancer are variable. In lung cancer, STAT3 can be activated by multiple pathways, including EGFR. Several approaches have been used to inhibit STAT3 in the hopes of developing an antitumor agent. Although several STAT3-specific agents are promising, none are in clinical development, mostly because of drug delivery and stability issues. In contrast, several JAK inhibitors are in clinical development. These orally available, ATP-competitive, small-molecule kinase inhibitors are being tested in myeloproliferative disorders. Future studies will determine whether JAK inhibitors are useful in the treatment of HNSCC or lung cancer.
Collapse
Affiliation(s)
- Stephen Y Lai
- Department of Head and Neck Surgery, The University of Texas M.D. Anderson Cancer Center at Houston, Houston, TX 77030, USA
| | | |
Collapse
|
42
|
Borghouts C, Tittmann H, Delis N, Kirchenbauer M, Brill B, Groner B. The intracellular delivery of a recombinant peptide derived from the acidic domain of PIAS3 inhibits STAT3 transactivation and induces tumor cell death. Mol Cancer Res 2010; 8:539-53. [PMID: 20371673 DOI: 10.1158/1541-7786.mcr-09-0417] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Signaling components, which confer an "addiction" phenotype on cancer cells, represent promising drug targets. The transcription factor signal transducers and activators of transcription 3 (STAT3) is constitutively activated in many different types of tumor cells and its activity is indispensible in a large fraction. We found that the expression of the endogenous inhibitor of STAT3, protein inhibitor of activated STAT3 (PIAS3), positively correlates with STAT3 activation in normal cells. This suggests that PIAS3 controls the extent and the duration of STAT3 activity in normal cells and thus prevents its oncogenic function. In cancer cells, however, the expression of PIAS3 is posttranscriptionally suppressed, possibly enhancing the oncogenic effects of activated STAT3. We delimited the interacting domains of STAT3 and PIAS3 and identified a short fragment of the COOH-terminal acidic region of PIAS3, which binds strongly to the coiled-coil domain of STAT3. This PIAS3 fragment was used to derive the recombinant STAT3-specific inhibitor rPP-C8. The addition of a protein transduction domain allowed the efficient internalization of rPP-C8 into cancer cells. This resulted in the suppression of STAT3 target gene expression, in the inhibition of migration and proliferation, and in the induction of apoptosis at low concentrations [half maximal effective concentration (EC(50)), <3 micromol/L]. rPP-C8 did not affect normal fibroblasts and represents an interesting lead for the development of novel cancer drugs targeting the coiled-coil domain of STAT3.
Collapse
Affiliation(s)
- Corina Borghouts
- Institute for Biomedical Research, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | | | | | | | | | | |
Collapse
|
43
|
Dabir S, Kluge A, Dowlati A. The association and nuclear translocation of the PIAS3-STAT3 complex is ligand and time dependent. Mol Cancer Res 2009; 7:1854-60. [PMID: 19903771 DOI: 10.1158/1541-7786.mcr-09-0313] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The epidermal growth factor (EGF) receptor activation of downstream signal transducers and activators of transcription 3 (STAT3) plays a crucial role in the pathogenesis of lung cancer. STAT3 transcriptional activity can be negatively regulated by protein inhibitor of activated STAT3 (PIAS3). We investigated the time-dependent PIAS3 shuffling and binding to STAT3 in an EGF-dependent model in lung cancer by using confocal microscopy, immunoprecipitation, luciferase reporter assay, and protein analysis of segregated cellular components. We also explored the role of phosphorylation at Tyr705 of STAT3 in the formation and intracellular shuffling of the PIAS3-STAT3 complex. In a growth factor-free state, PIAS3 was localized to the cytoplasm and unbound to STAT3 in both H520 and A549 cells. On exposure to EGF, we observed STAT3 phosphorylation and rapid formation of the PIAS3-STAT3 complex. Within 5 minutes, there was a progressive translocation of the complex to the nucleus, and by 10 minutes, PIAS3 was uniquely localized to the nuclear compartment. After 30 minutes, PIAS3 returned to the cytoplasm. Using site-directed mutagenesis, we substituted Tyr705 of STAT3 with a phenylalanine. Despite EGF stimulation, we observed a significant decrease in PIAS3-STAT3 binding and a significant reduction in nuclear translocation of PIAS3. Furthermore, there was a significant reduction in the capacity of PIAS3 to reduce STAT3-mediated gene transcription. In wild-type STAT3 cells, increasing concentrations of PIAS3 resulted in a proportional decrease in STAT3 phosphorylation. These data suggest an important role for the negative regulatory effect of PIAS3 on STAT3 in EGF-driven tumors.
Collapse
Affiliation(s)
- Snehal Dabir
- Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
44
|
Yu ZB, Bai L, Qian P, Xiao YB, Wang GS, Qian GS, Bai CX, Min JX. Restoration of SOCS3 suppresses human lung adenocarcinoma cell growth by downregulating activation of Erk1/2, Akt apart from STAT3. Cell Biol Int 2009; 33:995-1001. [PMID: 19524687 DOI: 10.1016/j.cellbi.2009.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Revised: 04/26/2009] [Accepted: 06/03/2009] [Indexed: 10/20/2022]
Abstract
SOCS3 is regarded as a major negative regulator of STAT3. Recent evidence indicates that SOCS3 regulates strength and duration of other signaling pathways including ras/ERK1/2/MAPK, PI3-K/Akt in non-malignant cells. The repression or silence of SOCS3 expression in a few tumor types has led to speculation that loss of SOCS3 gene is closely related to deregulation of multiple signal pathways during tumorigenesis. However, apart from STAT3, little is known in malignant cells about the mechanism by which SOCS3 modulates other intracellular signal cascades such as Erk1/2 and Akt, whose aberrant activation has been implicated in many human tumors. Expression of SOCS3 proved deficient in human lung adenocarcinoma A549 cells, and forced expression of SOCS3 resulted in growth inhibition. Growth suppression due to SOCS3 was associated with attenuated activation of Erk1/2, Akt as well as STAT3. The results suggested that SOCS3, as negative regulators of cytokine signaling, might maintain homeostasis by regulating multiple signaling pathways and reverse cell malignant behavior.
Collapse
Affiliation(s)
- Zu-Bin Yu
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, PR China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Neuwirt H, Puhr M, Santer FR, Susani M, Doppler W, Marcias G, Rauch V, Brugger M, Hobisch A, Kenner L, Culig Z. Suppressor of cytokine signaling (SOCS)-1 is expressed in human prostate cancer and exerts growth-inhibitory function through down-regulation of cyclins and cyclin-dependent kinases. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1921-30. [PMID: 19342366 PMCID: PMC2671279 DOI: 10.2353/ajpath.2009.080751] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 01/22/2009] [Indexed: 01/01/2023]
Abstract
Suppressor of cytokine signaling (SOCS) proteins play a pivotal role in the development and progression of various cancers. We have previously shown that SOCS-3 is expressed in prostate cancer, and its expression is inversely correlated with activation of signal transducer and activator of transcription factor 3. We hypothesized that SOCS-1, if expressed in prostate cancer cells, has a growth-regulatory role in this malignancy. The presence of both SOCS-1 mRNA and protein was detected in all tested cell lines. To assess SOCS-1 expression levels in vivo, we analyzed tissue microarrays and found a high percentage of positive cells in both prostate intraepithelial neoplasias and cancers. SOCS-1 expression levels decreased in samples taken from patients undergoing hormonal therapy but increased in specimens from patients who failed therapy. In LNCaP-interleukin-6- prostate cancer cells, SOCS-1 was up-regulated by interleukin-6 and in PC3-AR cells by androgens; such up-regulation was also found to significantly impair cell proliferation. To corroborate these findings, we used a specific small interfering RNA against SOCS-1 and blocked expression of the protein. Down-regulation of SOCS-1 expression caused a potent growth stimulation of PC3, DU-145, and LNCaP-interleukin-6- cells that was associated with the increased expression levels of cyclins D1 and E as well as cyclin-dependent kinases 2 and 4. In summary, we show that SOCS-1 is expressed in prostate cancer both in vitro and in vivo and acts as a negative growth regulator.
Collapse
Affiliation(s)
- Hannes Neuwirt
- Department of Urology, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Neoplasia: the second decade. Neoplasia 2009; 10:1314-24. [PMID: 19048110 DOI: 10.1593/neo.81372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 10/27/2008] [Accepted: 10/27/2008] [Indexed: 12/30/2022] Open
Abstract
This issue marks the end of the 10-year anniversary of Neoplasia where we have seen exciting growth in both number of submitted and published articles in Neoplasia. Neoplasia was first published in 1999. During the past 10 years, Neoplasia has dynamically adapted to the needs of the cancer research community as technologies have advanced. Neoplasia is currently providing access to articles through PubMed Central to continue to facilitate rapid broad-based dissemination of published findings to the scientific community through an Open Access model. This has in part helped Neoplasia to achieve an improved impact factor this past year, demonstrating that the manuscripts published by Neoplasia are of great interest to the overall cancer research community. This past year, Neoplasia received a record number of articles for review and has had a 21% increase in the number of published articles.
Collapse
|
47
|
Brantley EC, Nabors LB, Gillespie GY, Choi YH, Palmer CA, Harrison K, Roarty K, Benveniste EN. Loss of protein inhibitors of activated STAT-3 expression in glioblastoma multiforme tumors: implications for STAT-3 activation and gene expression. Clin Cancer Res 2008; 14:4694-704. [PMID: 18676737 DOI: 10.1158/1078-0432.ccr-08-0618] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE STATs activate transcription in response to numerous cytokines, controlling proliferation, gene expression, and apoptosis. Aberrant activation of STAT proteins, particularly STAT-3, is implicated in the pathogenesis of many cancers, including GBM, by promoting cell cycle progression, stimulating angiogenesis, and impairing tumor immune surveillance. Little is known about the endogenous STAT inhibitors, the PIAS proteins, in human malignancies. The objective of this study was to examine the expression of STAT-3 and its negative regulator, PIAS3, in human tissue samples from control and GBM brains. EXPERIMENTAL DESIGN Control and GBM human tissues were analyzed by immunoblotting and immunohistochemistry to determine the activation status of STAT-3 and expression of the PIAS3 protein. The functional consequence of PIAS3 inhibition by small interfering RNA or PIAS3 overexpression in GBM cells was determined by examining cell proliferation, STAT-3 transcriptional activity, and STAT-3 target gene expression. This was accomplished using [(3)H]TdR incorporation, STAT-3 dominant-negative constructs, reverse transcription-PCR, and immunoblotting. RESULTS AND CONCLUSIONS STAT-3 activation, as assessed by tyrosine and serine phosphorylation, was elevated in GBM tissue compared with control tissue. Interestingly, we observed expression of PIAS3 in control tissue, whereas PIAS3 protein expression in GBM tissue was greatly reduced. Inhibition of PIAS3 resulted in enhanced glioblastoma cellular proliferation. Conversely, PIAS3 overexpression inhibited STAT-3 transcriptional activity, expression of STAT-3-regulated genes, and cell proliferation. We propose that the loss of PIAS3 in GBM contributes to enhanced STAT-3 transcriptional activity and subsequent cell proliferation.
Collapse
Affiliation(s)
- Emily C Brantley
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294-0005, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Ghoshal (Gupta) S, Baumann H, Wetzler M. Epigenetic regulation of signal transducer and activator of transcription 3 in acute myeloid leukemia. Leuk Res 2008; 32:1005-14. [PMID: 18192010 PMCID: PMC4629448 DOI: 10.1016/j.leukres.2007.11.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Revised: 11/25/2007] [Accepted: 11/26/2007] [Indexed: 12/22/2022]
Abstract
We have demonstrated that constitutive signal transducer and activator of transcription (STAT) 3 activity, observed in approximately 50% of acute myeloid leukemia (AML) cases, is associated with adverse treatment outcome. Constitutive STAT3 activation may result from the expression of oncogenic protein tyrosine kinases or from autocrine stimulation by hematopoietic growth factors. These causes are generally neither necessary nor sufficient for leukemogenesis; additional transforming events or growth stimulatory processes are needed. Here we review the literature addressing epigenetic regulation as a mechanism controlling STAT3 signaling in AML. A better understanding of mechanisms of dysregulation of STAT signaling pathways may serve as a basis for designing novel therapeutic strategies that target these pathways in leukemia cells.
Collapse
Affiliation(s)
- Sampa Ghoshal (Gupta)
- Departments of Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263
| | - Heinz Baumann
- Molecular and Cellular Biology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263
| | - Meir Wetzler
- Departments of Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263
| |
Collapse
|
49
|
Brantley EC, Benveniste EN. Signal transducer and activator of transcription-3: a molecular hub for signaling pathways in gliomas. Mol Cancer Res 2008; 6:675-84. [PMID: 18505913 PMCID: PMC3886801 DOI: 10.1158/1541-7786.mcr-07-2180] [Citation(s) in RCA: 176] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Glioblastoma is the most common and severe primary brain tumor in adults. Its aggressive and infiltrative nature renders the current therapeutics of surgical resection, radiation, and chemotherapy relatively ineffective. Accordingly, recent research has focused on the elucidation of various signal transduction pathways in glioblastoma, particularly aberrant activation. This review focuses on the signal transducer and activator of transcription-3 (STAT-3) signal transduction pathway in the context of this devastating tumor. STAT-3 is aberrantly activated in human glioblastoma tissues, and this activation is implicated in controlling critical cellular events thought to be involved in gliomagenesis, such as cell cycle progression, apoptosis, angiogenesis, and immune evasion. There are no reports of gain-of-function mutations in glioblastoma; rather, the activation of STAT-3 is thought to be a consequence of either dysregulation of upstream kinases or loss of endogenous inhibitors. This review provides detailed insight into the multiple mechanisms of STAT-3 activation in glioblastoma, as well as describing endogenous and chemical inhibitors of this pathway and their clinical significance. In glioblastoma, STAT-3 acts a molecular hub to link extracellular signals to transcriptional control of proliferation, cell cycle progression, and immune evasion. Because STAT-3 plays this central role in glioblastoma signal transduction, it has significant potential as a therapeutic target.
Collapse
Affiliation(s)
- Emily C Brantley
- Department of Cell Biology, 1918 University Boulevard, MCLM 395A, University of Alabama at Birmingham, Birmingham, AL 35294-0005, USA
| | | |
Collapse
|
50
|
Fujita M, Zhu X, Sasaki K, Ueda R, Low KL, Pollack IF, Okada H. Inhibition of STAT3 promotes the efficacy of adoptive transfer therapy using type-1 CTLs by modulation of the immunological microenvironment in a murine intracranial glioma. THE JOURNAL OF IMMUNOLOGY 2008; 180:2089-98. [PMID: 18250414 DOI: 10.4049/jimmunol.180.4.2089] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A variety of cancers, including malignant gliomas, show aberrant activation of STAT3, which plays a pivotal role in negative regulation of antitumor immunity. We hypothesized that inhibition of STAT3 signals would improve the efficacy of T cell adoptive transfer therapy by reversal of STAT3-induced immunosuppression in a murine GL261 intracranial glioma model. In vitro treatment of GL261 cells with JSI-124, a STAT3 inhibitor, reversed highly phosphorylated status of STAT3. Systemic i.p. administration of JSI-124 in glioma-bearing immunocompetent mice, but not athymic mice, resulted in prolonged survival, suggesting a role of adaptive immunity in the antitumor effect. Furthermore, JSI-124 promoted maturation of tumor-infiltrating CD11c(+) dendritic cells and activation of tumor-conditioned cytotoxic T cells, enhanced dendritic cells and GL261 production of CXCL-10, a critical chemokine for attraction of Tc1 cells. When i.p. JSI-124 administration was combined with i.v. transfer of Pmel-I mouse-derived type-1 CTLs (Tc1), glioma-bearing mice exhibited prolonged survival compared with i.p. JSI-124 or i.v. Tc1 therapy alone. Flow cytometric analyses of brain infiltrating lymphocytes revealed that JSI-124-treatment enhanced the tumor-homing of i.v. transferred Tc1 cells in a CXCL-10-dependent fashion. Systemic JSI-124 administration also up-regulated serum IL-15 levels, and promoted the persistence of transferred Tc1 in the host. These data suggest that systemic inhibition of STAT3 signaling can reverse the suppressive immunological environment of intracranial tumor bearing mice both systemically and locally, thereby promoting the efficacy of adoptive transfer therapy with Tc1.
Collapse
Affiliation(s)
- Mitsugu Fujita
- Department of Neurological Surgery and Brain Tumor Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15232, USA
| | | | | | | | | | | | | |
Collapse
|