1
|
Kim Y, Jeon E, Ahn H, Kang J, Sim T. Identification of Thieno[3,2-d]pyrimidine derivatives as potent and selective Janus Kinase 1 inhibitors. Eur J Med Chem 2025; 286:117308. [PMID: 39892337 DOI: 10.1016/j.ejmech.2025.117308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/30/2024] [Accepted: 01/11/2025] [Indexed: 02/03/2025]
Abstract
Being a primary driver in oncogenic activations of JAK-STAT signaling pathway, Janus Kinase 1 (JAK1) stands out as a promising target in anti-cancer drug discovery. We employed a scaffold morphing strategy to design and synthesize thieno[3,2-d]pyrimidine derivatives, which led to identification of 24 as a potent and highly selective JAK1 inhibitor. Kinome-wide selectivity profiling reveals that 24 exhibits a high degree of selectivity for JAK1 among the 370 kinases tested. SAR study demonstrates that both 25 and 46, improved derivatives of 24, possess higher selectivity towards JAK1 over JAK2 and JAK3 compared to AZD4205 (9). It is of note that 46 has 4-fold higher enzymatic activity against JAK1 (IC50 = 0.022 μM) relative to 9. Moreover, both 25 and 46 demonstrate over 5-fold enhancement in anti-proliferative activities on NSCLC cells with regard to 9, accompanied by significant inhibition of JAK1 signaling. Compared with 9, derivative 24, 25, and 46 induce more strongly apoptosis, cell cycle arrest, and reduction of colony formation on NSCLC cells. Our findings offer valuable insights into the design of novel selective JAK1 inhibitors.
Collapse
Affiliation(s)
- Younghoon Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Department of Biomedical Sciences, Graduate School of Medical Science, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Eunhye Jeon
- Department of Biomedical Sciences, Graduate School of Medical Science, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyunwoo Ahn
- Graduate School of Clinical Drug Discovery & Development, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Juhee Kang
- Graduate School of Clinical Drug Discovery & Development, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Taebo Sim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Department of Biomedical Sciences, Graduate School of Medical Science, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; Graduate School of Clinical Drug Discovery & Development, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; Clinical Candidate Discovery & Development Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; Department of Medical Science, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
2
|
Mathews SG, Krishna RD, M. L, K. N, Murali S, Agarwal P, Rani E, F. AM. The Role of the Plasminogen Activator Inhibitor 1 ( PAI1 ) in Ovarian Cancer: Mechanisms and Therapeutic Implications. Glob Med Genet 2024; 11:358-365. [PMID: 39583124 PMCID: PMC11521755 DOI: 10.1055/s-0044-1791734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024] Open
Abstract
Ovarian cancer (OC) is one among most significantly fatal gynecological cancers, with late-stage detection and an inadequate prognosis. Plasminogen activator inhibitor-1 ( PAI1 ) gene anticipates negative outcomes in many different kinds of malignancies. Several research investigations are currently being done to examine the biological role of PAI1 in OC and the possible benefits of targeted pharmacotherapies. The PAI1 gene has been linked to the emergence and development of cancer in the ovary. PAI1 , an inhibitor of serine protease, influences the fibrinolysis and extracellular matrix remodeling, both of which are crucial for tumor expansion and metastatic growth. PAI1 levels have been discovered to be subsequently more elevated in malignant ovarian tissues than in usual ovarian tissue, demonstrating a potential connection among PAI1 overexpression and OC development. PAI1 promotes tumor cell proliferation, movement, and an invasion by influencing the urokinase-plasminogen activators and through interactions with cell surface receptors. In addition, PAI1 gene contributes to angiogenesis and apoptotic cell death, which contribute to the more hostile phenotypes of OC. The prognostic and therapeutic consequences of focusing on PAI1 in OC are explored, demonstrating PAI1 's potential to be a biomarker and emphasizing for novel treatment approaches. The PAI1 gene possesses several functions in OC, affecting tumor development, an invasion, and metastatic growth. Comprehending the complicated interactions and mechanisms that regulate PAI1 in OC may lead to more efficient evaluation and treatment strategies and ultimately enhance patient outcomes.
Collapse
Affiliation(s)
- Sneha Grace Mathews
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - R.B. Devi Krishna
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Lavanya M.
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Nandini K.
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Sanjana Murali
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Preet Agarwal
- Department of Gynecology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Elizabeth Rani
- Department of Biotechnology, Hindustan College of Arts and Science, Chennai, Tamil Nadu, India
| | - Andrea Mary F.
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| |
Collapse
|
3
|
Rodman EPB, Emch MJ, Hou X, Bajaj A, Pearson NA, John AJ, Ortiz Y, Bass AD, Singh S, Baldassarre G, Kaufmann SH, Weroha SJ, Hawse JR. Lestaurtinib's antineoplastic activity converges on JAK/STAT signaling to inhibit advanced forms of therapy resistant ovarian cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597753. [PMID: 38895264 PMCID: PMC11185641 DOI: 10.1101/2024.06.06.597753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Ovarian cancer is the deadliest gynecological malignancy, owing to its late-stage diagnosis and high rates of recurrence and resistance following standard-of-care treatment, highlighting the need for novel treatment approaches. Through an unbiased drug screen, we identified the kinase inhibitor, lestaurtinib, as a potent antineoplastic agent for chemotherapy- and PARP-inhibitor (PARPi)-sensitive and -resistant ovarian cancer cells and patient derived xenografts (PDXs). RNA-sequencing revealed that lestaurtinib potently suppressed JAK/STAT signaling and lestaurtinib efficacy was shown to be directly related to JAK/STAT pathway activity in cell lines and PDX models. Most ovarian cancer cells exhibited constitutive JAK/STAT pathway activation and genetic loss of STAT1 and STAT3 resulted in growth inhibition. Lestaurtinib also displayed synergy when combined with cisplatin and olaparib, including in a model of PARPi resistance. In contrast, the most well-known JAK/STAT inhibitor, ruxolitinib, lacked antineoplastic activity against all ovarian cancer cell lines and PDX models tested. This divergent behavior was reflected in the ability of lestaurtinib to block both Y701/705 and S727 phosphorylation of STAT1 and STAT3, whereas ruxolitinib failed to block S727. Consistent with these findings, lestaurtinib additionally inhibited JNK and ERK activity, leading to more complete suppression of STAT phosphorylation. Concordantly, combinatorial treatment with ruxolitinib and a JNK or ERK inhibitor resulted in synergistic antineoplastic effects at dose levels where single agents were ineffective. Taken together, these findings indicate that lestaurtinib, and other treatments that converge on JAK/STAT signaling, are worthy of further pre-clinical and clinical exploration for the treatment of highly aggressive and advanced forms of ovarian cancer. Statement of significance Lestaurtinib is a novel inhibitor of ovarian cancer, including chemotherapy- and PARPi-resistant models, that acts through robust inhibition of the JAK/STAT pathway and synergizes with standard-of-care agents at clinically relevant concentrations.
Collapse
|
4
|
Yunianto I, Currie M, Chitcholtan K, Sykes P. Potential drug repurposing of ruxolitinib to inhibit the JAK/STAT pathway for the treatment of patients with epithelial ovarian cancer. J Obstet Gynaecol Res 2023; 49:2563-2574. [PMID: 37565583 DOI: 10.1111/jog.15761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 07/25/2023] [Indexed: 08/12/2023]
Abstract
AIM This review aimed to describe the potential for therapeutic targeting of the JAK/STAT signaling pathway by repurposing the clinically-approved JAK inhibitor ruxolitinib in the patients with epithelial ovarian cancer (OC) setting. METHODS We reviewed publications that focus on the inhibition of the JAK/STAT pathway in hematological and solid malignancies including OC. RESULTS Preclinical studies showed that ruxolitinib effectively reduces OC cell viability and metastasis and enhances the anti-tumor activity of chemotherapy drugs. There are a number of recent clinical trials exploring the role of JAK/STAT inhibition in solid cancers including OC. Early results have not adequately supported efficacy in solid tumors. However, there are preclinical data and clinical studies supporting the use of ruxolitinib in combination with both chemotherapy and other targeted drugs in OC setting. CONCLUSION Inflammatory conditions and persistent activation of the JAK/STAT pathway are associated with tumourigenesis and chemoresistance, and therapeutic blockade of this pathway shows promising results. For women with OC, clinical investigation exploring the role of ruxolitinib in combination with chemotherapy agents or other targeted therapeutics is warranted.
Collapse
Affiliation(s)
- Irfan Yunianto
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
- Department of Biology Education, Universitas Ahmad Dahlan, Indonesia
| | - Margaret Currie
- Department of Pathology and Biomedical Sciences, University of Otago, Christchurch, New Zealand
| | - Kenny Chitcholtan
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| | - Peter Sykes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| |
Collapse
|
5
|
Standing D, Feess E, Kodiyalam S, Kuehn M, Hamel Z, Johnson J, Thomas SM, Anant S. The Role of STATs in Ovarian Cancer: Exploring Their Potential for Therapy. Cancers (Basel) 2023; 15:2485. [PMID: 37173951 PMCID: PMC10177275 DOI: 10.3390/cancers15092485] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Ovarian cancer (OvCa) is a deadly gynecologic malignancy that presents many clinical challenges due to late-stage diagnoses and the development of acquired resistance to standard-of-care treatment protocols. There is an increasing body of evidence suggesting that STATs may play a critical role in OvCa progression, resistance, and disease recurrence, and thus we sought to compile a comprehensive review to summarize the current state of knowledge on the topic. We have examined peer reviewed literature to delineate the role of STATs in both cancer cells and cells within the tumor microenvironment. In addition to summarizing the current knowledge of STAT biology in OvCa, we have also examined the capacity of small molecule inhibitor development to target specific STATs and progress toward clinical applications. From our research, the best studied and targeted factors are STAT3 and STAT5, which has resulted in the development of several inhibitors that are under current evaluation in clinical trials. There remain gaps in understanding the role of STAT1, STAT2, STAT4, and STAT6, due to limited reports in the current literature; as such, further studies to establish their implications in OvCa are necessitated. Moreover, due to the deficiency in our understanding of these STATs, selective inhibitors also remain elusive, and therefore present opportunities for discovery.
Collapse
Affiliation(s)
- David Standing
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Emma Feess
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Satvik Kodiyalam
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Michael Kuehn
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Zachary Hamel
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Jaimie Johnson
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Sufi Mary Thomas
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Shrikant Anant
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
6
|
Seo J, Lee DE, Kim SM, Kim E, Kim JK. Licochalcone A Exerts Anti-Cancer Activity by Inhibiting STAT3 in SKOV3 Human Ovarian Cancer Cells. Biomedicines 2023; 11:biomedicines11051264. [PMID: 37238935 DOI: 10.3390/biomedicines11051264] [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: 02/23/2023] [Revised: 03/31/2023] [Accepted: 04/18/2023] [Indexed: 05/28/2023] Open
Abstract
Licochalcone A (LicA), a major active component of licorice, has been reported to exhibit various pharmacological actions. The purpose of this study was to investigate the anticancer activity of LicA and detail its molecular mechanisms against ovarian cancer. SKOV3 human ovarian cancer cells were used in this study. Cell viability was measured using a cell counting kit-8 assay. The percentages of apoptotic cells and cell cycle arrest were determined by flow cytometry and Muse flow cytometry. The expression levels of proteins regulating cell apoptosis, cell cycle, and the signal transducer and activator of transcription 3 (STAT3) signaling pathways were examined using Western blotting analysis. The results indicated that LicA treatment inhibited the cell viability of SKOV3 cells and induced G2/M phase arrest. Furthermore, LicA induced an increase in ROS levels, a reduction in mitochondrial membrane potential, and apoptosis accompanied by an increase in cleaved caspases and cytoplasmic cytochrome c. Additionally, LicA caused a dramatic decrease in STAT3 protein levels, but not mRNA levels, in SKOV3 cells. Treatment with LicA also reduced phosphorylation of the mammalian target of rapamycin and eukaryotic translation initiation factor 4E-binding protein in SKOV3 cells. The anti-cancer effects of LicA on SKOV3 cells might be mediated by reduced STAT3 translation and activation.
Collapse
Affiliation(s)
- Jeonghyeon Seo
- Department of Biomedical Science, Daegu Catholic University, Gyeongsan-si 38430, Republic of Korea
| | - Da Eun Lee
- Department of Biomedical Science, Daegu Catholic University, Gyeongsan-si 38430, Republic of Korea
| | - Seong Mi Kim
- Department of Biomedical Science, Daegu Catholic University, Gyeongsan-si 38430, Republic of Korea
| | - Eunjung Kim
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan-si 38430, Republic of Korea
| | - Jin-Kyung Kim
- Department of Biomedical Science, Daegu Catholic University, Gyeongsan-si 38430, Republic of Korea
| |
Collapse
|
7
|
Agrawal MY, Gaikwad S, Srivastava S, Srivastava SK. Research Trend and Detailed Insights into the Molecular Mechanisms of Food Bioactive Compounds against Cancer: A Comprehensive Review with Special Emphasis on Probiotics. Cancers (Basel) 2022; 14:cancers14225482. [PMID: 36428575 PMCID: PMC9688469 DOI: 10.3390/cancers14225482] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/26/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
In an attempt to find a potential cure for cancer, scientists have been probing the efficacy of the food we eat and its bioactive components. Over the decades, there has been an exponentially increasing trend of research correlating food and cancer. This review explains the molecular mechanisms by which bioactive food components exhibit anticancer effects in several cancer models. These bioactive compounds are mainly plant based or microbiome based. While plants remain the primary source of these phytochemicals, little is known about probiotics, i.e., microbiome sources, and their relationships with cancer. Thus, the molecular mechanisms underlying the anticancer effect of probiotics are discussed in this review. The principal mode of cell death for most food bioactives is found to be apoptosis. Principal oncogenic signaling axes such as Akt/PI3K, JAK/STAT, and NF-κB seem to be modulated due to these bioactives along with certain novel targets that provide a platform for further oncogenic research. It has been observed that probiotics have an immunomodulatory effect leading to their chemopreventive actions. Various foods exhibit better efficacy as complete extracts than their individual phytochemicals, indicating an orchestrated effect of the food components. Combining bioactive agents with available chemotherapies helps synergize the anticancer action of both to overcome drug resistance. Novel techniques to deliver bioactive agents enhance their therapeutic response. Such combinations and novel approaches are also discussed in this review. Notably, most of the food components that have been studied for cancer have shown their efficacy in vivo. This bolsters the claims of these studies and, thus, provides us with hope of discovering anticancer agents in the food that we eat.
Collapse
Affiliation(s)
- Manas Yogendra Agrawal
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
- Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Shreyas Gaikwad
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
- Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | | | - Sanjay K. Srivastava
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
- Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
- Correspondence: ; Tel.: +1-325-696-0464; Fax: +1-325-676-3875
| |
Collapse
|
8
|
Oliveira MM, Bonturi CR, Salu BR, Oliva MLV, Mortara RA, Orikaza CM. Modulation of STAT-1, STAT-3, and STAT-6 activities in THP-1 derived macrophages infected with two Trypanosoma cruzi strains. Front Immunol 2022; 13:1038332. [PMID: 36389843 PMCID: PMC9643828 DOI: 10.3389/fimmu.2022.1038332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/04/2022] [Indexed: 12/03/2022] Open
Abstract
Trypanosoma cruzi is the causative protozoan of Chagas' Disease, a neglected tropical disease that affects 6-7 million people worldwide. Interaction of the parasite with the host immune system is a key factor in disease progression and chronic symptoms. Although the human immune system is capable of controlling the disease, the parasite has numerous evasion mechanisms that aim to maintain intracellular persistence and survival. Due to the pronounced genetic variability of T. cruzi, co-infections or mixed infections with more than one parasite strain have been reported in the literature. The intermodulation in such cases is unclear. This study aimed to evaluate the co-infection of T. cruzi strains G and CL compared to their individual infections in human macrophages derived from THP-1 cells activated by classical or alternative pathways. Flow cytometry analysis demonstrated that trypomastigotes were more infective than extracellular amastigotes (EAs) and that strain G could infect more macrophages than strain CL. Classically activated macrophages showed lower number of infected cells and IL-4-stimulated cells displayed increased CL-infected macrophages. However, co-infection was a rare event. CL EAs decreased the production of reactive oxygen species (ROS), whereas G trypomastigotes displayed increased ROS detection in classically activated cells. Co-infection did not affect ROS production. Monoinfection by strain G or CL mainly induced an anti-inflammatory cytokine profile by decreasing inflammatory cytokines (IFN-γ, TNF-α, IL-1β) and/or increasing IL-4, IL-10, and TGF-β. Co-infection led to a predominant inflammatory milieu, with reduced IL-10 and TGF-β, and/or promotion of IFN-γ and IL-1β release. Infection by strain G reduced activation of intracellular signal transducer and activator of transcription (STAT) factors. In EAs, monoinfections impaired STAT-1 activity and promoted phosphorylation of STAT-3, both changes may prolong cell survival. Coinfected macrophages displayed pronounced activation of all STATs examined. These activations likely promoted parasite persistence and survival of infected cells. The collective results demonstrate that although macrophages respond to both strains, T. cruzi can modulate the intracellular environment, inducing different responses depending on the strain, parasite infective form, and co-infection or monoinfection. The modulation influences parasite persistence and survival of infected cells.
Collapse
Affiliation(s)
- Melissa Martins Oliveira
- ¹Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Camila Ramalho Bonturi
- ²Biochemistry Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Bruno Ramos Salu
- ²Biochemistry Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Maria Luiza Vilela Oliva
- ²Biochemistry Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Renato Arruda Mortara
- ¹Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Cristina Mary Orikaza
- ¹Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| |
Collapse
|
9
|
Jia L, Huang P, Lin T, Lin C, Ding X, Lin L, Zhu L, Zhou Z. CircPUM1 Knockdown Confers Radiosensitivity in Oral Squamous Cell Carcinoma by Regulating the miR-580/STAT3 Pathway. Front Genet 2022; 13:907219. [PMID: 36092922 PMCID: PMC9452959 DOI: 10.3389/fgene.2022.907219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/20/2022] [Indexed: 11/20/2022] Open
Abstract
Background: CircPUM1 acts as an oncogene in a variety of tumors, and there is no related research on oral squamous cell carcinoma. This study aimed to evaluate the clinical significance of CircPUM1 in oral squamous cell carcinoma radiotherapy. Methods: Radio-resistant cell lines were established by increasing the X-ray dose. Analysis of CircPUM1 expression in oral squamous cell carcinoma was carried out using bioinformatics tools. Cell proliferation was analyzed with CCK-8 and colony formation. Protein and gene expressions were detected by Western blotting and qPCR. RNA interference inhibits endogenous gene expression. A luciferase reporter system and immunoprecipitation were used to validate the target of CircPUM1. Result: CircPUM1 was highly expressed in OSCC. The higher the expression level of CircPUM1 in OSCC, the worse the clinical features and prognosis. Knockdown of CircPUM1 enhances the sensitivity of OSCC cells to X-rays, and expression of exogenous CircPUM1 makes OSCC cells acquire radiation resistance. The absence of CircPUM1 blocked the cells in the G0/G1 phase and triggered apoptosis. The prediction of mir-580-binding site, luciferase reporter system, and immunoprecipitation confirmed that mir-580 is the binding site of CircPUM1. In addition, STAT3 was predicted and confirmed as the binding site of mir-580. Overexpression of STAT3 partially attenuated the radiosensitivity of OSCC cells to knockdown of CircPUM1. Conclusion: CircPUM1 has the oncogene expression profile in oral squamous cell carcinoma; patients with high expression of CircPUM1 have less benefit from radiotherapy and need more frequent follow-up. In addition, CircPUM1 may be a potential therapeutic target for oral squamous cell carcinoma. The CircPUM1/mir-580/STAT3 axis has a certain effect on the radiosensitivity of OSCC. These results suggest that patients with low expression of CircPUM1 may gain more benefits.
Collapse
Affiliation(s)
- Linghui Jia
- Fujian Key Laboratory of Oral Diseases, Fujian Provincial Engineering Research Center of Oral Biomaterial of Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Department of Prosthodontics, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Pengcheng Huang
- Fujian Key Laboratory of Oral Diseases, Fujian Provincial Engineering Research Center of Oral Biomaterial of Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Department of Orthodontics, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Tingting Lin
- Affiliated Sanming First Hospital, Fujian Medical University, Sanming, China
| | - Chunyan Lin
- Affiliated Sanming First Hospital, Fujian Medical University, Sanming, China
| | - Xiaofen Ding
- Affiliated Sanming First Hospital, Fujian Medical University, Sanming, China
| | - Liping Lin
- Affiliated Sanming First Hospital, Fujian Medical University, Sanming, China
| | - Lifeng Zhu
- Affiliated Sanming First Hospital, Fujian Medical University, Sanming, China
- *Correspondence: Zhilian Zhou, ; Lifeng Zhu,
| | - Zhilian Zhou
- Affiliated Sanming First Hospital, Fujian Medical University, Sanming, China
- *Correspondence: Zhilian Zhou, ; Lifeng Zhu,
| |
Collapse
|
10
|
Zhang T, Li S, Li J, Yin F, Hua Y, Wang Z, Wang H, Zuo D, Xu J, Cai Z. Pectolinarigenin acts as a potential anti-osteosarcoma agent via mediating SHP-1/JAK2/STAT3 signaling. Biomed Pharmacother 2022; 153:113323. [PMID: 35752008 DOI: 10.1016/j.biopha.2022.113323] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/10/2022] [Accepted: 06/20/2022] [Indexed: 11/28/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) plays essential roles in cancer progression and has been considered as a promising target for cancer therapy. Here, we used a dual luciferase assay to identify that pectolinarigenin inhibited STAT3 transcriptional activity. Further, results showed pectolinarigenin inhibited constitutive and IL6 induced STAT3 signaling, diminished the accumulation of STAT3 in the nucleus, dimerization and blocked STAT3 DNA binding activity. Mechanism investigations indicated that pectolinarigenin disturbed the STAT3/DNMT1/HDAC1 complex formation in the promoter region of SHP-1, which reversely mediates STAT3 signaling, leading to the upregulation of SHP-1 expression in osteosarcoma. We also found pectolinarigenin significantly suppressed osteosarcoma growth, induced apoptosis. In addition, pectolinarigenin blocked tumor cells migration, invasion and reserved EMT phenotype. In spontaneous tibial injection and patient-derived xenograft models of osteosarcoma, we identified administration (i.p.) of pectolinarigenin (20 mg/kg/2 days and 50 mg/kg/2 days) blocked STAT3 activation and disturbed tumor growth and metastasis with superior pharmacodynamic properties. Taken together, our findings demonstrate that pectolinarigenin may be a candidate for osteosarcoma intervention linked to its STAT3 signaling inhibitory activity.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China.
| | - Suoyuan Li
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China; Suzhou Municipal Hospital, Suzhou, PR China
| | - Jingjie Li
- Translational Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Fei Yin
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Yingqi Hua
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Zhuoying Wang
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Hongsheng Wang
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Dongqing Zuo
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Jing Xu
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Zhengdong Cai
- Department of Orthopedics, Shanghai Bone Tumor Institution, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China.
| |
Collapse
|
11
|
Mahata S, Behera SK, Kumar S, Sahoo PK, Sarkar S, Fazil MHUT, Nasare VD. In-silico and in-vitro investigation of STAT3-PIM1 heterodimeric complex: Its mechanism and inhibition by curcumin for cancer therapeutics. Int J Biol Macromol 2022; 208:356-366. [DOI: 10.1016/j.ijbiomac.2022.03.137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/19/2022] [Accepted: 03/22/2022] [Indexed: 01/19/2023]
|
12
|
Park SA, Kim LK, Park HM, Kim HJ, Heo TH. Inhibition of GP130/STAT3 and EMT by combined bazedoxifene and paclitaxel treatment in ovarian cancer. Oncol Rep 2022; 47:52. [PMID: 35029286 PMCID: PMC8771159 DOI: 10.3892/or.2022.8263] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023] Open
Abstract
The interleukin 6 (IL‑6)/glycoprotein 130 (GP130)/signal transducer and activator of transcription 3 (STAT3) signalling pathway, with GP130 as an intermediate membrane receptor, is involved in the survival, metastasis, and resistance of ovarian cancer. Bazedoxifene, an FDA‑approved drug, is an inhibitor of GP130 and a selective estrogen modulator (SERM). We studied the mechanism of the combination therapy of bazedoxifene and paclitaxel in inhibiting the IL‑6‑mediated GP130/STAT3 signaling pathway in ovarian cancer. Surface plasmon resonance (SPR) was used to assess the binding of bazedoxifene to GP130. Migration, invasion, and apoptosis of ovarian cancer cells were assessed using bazedoxifene and paclitaxel. In addition, we determined the effects of bazedoxifene and paclitaxel alone or in combination on the GP130/STAT3 pathway and epithelial‑mesenchymal transition (EMT). The results revealed that the combination of bazedoxifene and paclitaxel suppressed cell viability, migration, and invasion in the ovarian cancer cells. In addition, the combination treatment increased apoptosis. Furthermore, bazedoxifene combined with paclitaxel inhibited the growth of ovarian cancer cells in a xenograft tumour model. This combination reduced STAT3 phosphorylation and suppressed gene expression and EMT. In conclusion, inhibition of GP130/STAT3 signalling and EMT via a combination of bazedoxifene and paclitaxel could be used as a therapeutic strategy by which to overcome ovarian cancer.
Collapse
Affiliation(s)
- Sun-Ae Park
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Lee Kyung Kim
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Hye Min Park
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Hee Jung Kim
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Tae-Hwe Heo
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| |
Collapse
|
13
|
Mitra S, Anand U, Jha NK, Shekhawat MS, Saha SC, Nongdam P, Rengasamy KRR, Proćków J, Dey A. Anticancer Applications and Pharmacological Properties of Piperidine and Piperine: A Comprehensive Review on Molecular Mechanisms and Therapeutic Perspectives. Front Pharmacol 2022; 12:772418. [PMID: 35069196 PMCID: PMC8776707 DOI: 10.3389/fphar.2021.772418] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/18/2021] [Indexed: 12/26/2022] Open
Abstract
Piperine and piperidine are the two major alkaloids extracted from black pepper (Piper nigrum); piperidine is a heterocyclic moiety that has the molecular formula (CH2)5NH. Over the years, many therapeutic properties including anticancer potential of these two compounds have been observed. Piperine has therapeutic potential against cancers such as breast cancer, ovarian cancer, gastric cancer, gliomal cancer, lung cancer, oral squamous, chronic pancreatitis, prostate cancer, rectal cancer, cervical cancer, and leukemia. Whereas, piperidine acts as a potential clinical agent against cancers, such as breast cancer, prostate cancer, colon cancer, lung cancer, and ovarian cancer, when treated alone or in combination with some novel drugs. Several crucial signalling pathways essential for the establishment of cancers such as STAT-3, NF-κB, PI3k/Aκt, JNK/p38-MAPK, TGF-ß/SMAD, Smac/DIABLO, p-IκB etc., are regulated by these two phytochemicals. Both of these phytochemicals lead to inhibition of cell migration and help in cell cycle arrest to inhibit survivability of cancer cells. The current review highlights the pharmaceutical relevance of both piperine and piperidine against different types of cancers.
Collapse
Affiliation(s)
- Sicon Mitra
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Uttpal Anand
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Mahipal S Shekhawat
- Department of Plant Biology and Biotechnology, Kanchi Mamunivar Government Institute for Postgraduate Studies and Research, Lawspet, India
| | - Suchismita Chatterjee Saha
- Department of Zoology, Nabadwip Vidyasagar College (Affiliated to the University of Kalyani), Nabadwip, India
| | | | - Kannan R R Rengasamy
- Green Biotechnologies Research Centre of Excellence, University of Limpopo, Sovenga, South Africa
| | - Jarosław Proćków
- Department of Plant Biology, Institute of Environmental Biology, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Abhijit Dey
- Ethnopharmacology and Natural Product Research Laboratory, Department of Life Sciences, Presidency University, Kolkata, India
| |
Collapse
|
14
|
Wu W, Yu F, Yu N, Zhu Y, Wu W, Gao P, Chen C. Glucose-6-phosphate dehydrogenase promotes the proliferation and migration of lung adenocarcinoma cells via the STAT3 signaling pathway. J Mol Histol 2022; 53:215-225. [PMID: 35028787 DOI: 10.1007/s10735-021-10045-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 12/01/2021] [Indexed: 10/19/2022]
Abstract
Lung adenocarcinoma (LUAD) is the most common histological subtype of lung cancer, and the leading cause of cancer-related deaths worldwide. G6PD has been reported to enhance the progression of various tumors by regulating the intracellular redox state and mediating nucleic acid synthesis. However, the biological role and molecular mechanism of G6PD in LUAD remain largely unknown. In this study, we found that G6PD was significantly upregulated in LUAD specimens and cell lines, and that the high levels of G6PD expression were closely associated with a poor prognosis for LUAD patients. Moreover, we found that G6PD significantly promoted the proliferation and migration of LUAD cells in vitro, and overexpression of G6PD also play a role of facilitating tumorigenesis in in vivo experiments. Mechanistically, the STAT3 signaling pathway was significantly activated by G6PD-mediated LUAD progression. Overall, our results suggest that G6PD could serve as a novel prognostic marker and therapeutic target for treating LUAD.
Collapse
Affiliation(s)
- Weidong Wu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China.,Fujian Provincial Key Laboratory of Cardiothoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Fengqiang Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Nanding Yu
- Department of Respiratory Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yong Zhu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Weihan Wu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Pengqiang Gao
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chun Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China. .,Fujian Provincial Key Laboratory of Cardiothoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China. .,Department of Thoracic Surgery, Fujian Medical University Union Hospital, #29 Xinquan Road, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
15
|
Li WZ, Xi HZ, Wang YJ, Ma HB, Cheng ZQ, Yang Y, Wu ML, Liu TM, Yang W, Wang Q, Liao MY, Xia Y, Zhang YW. Design, synthesis, and biological evaluation of benzo[b]thiophene 1,1-dioxide derivatives as potent STAT3 inhibitors. Chem Biol Drug Des 2021; 98:835-849. [PMID: 34416096 DOI: 10.1111/cbdd.13939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/06/2021] [Accepted: 07/23/2021] [Indexed: 02/05/2023]
Abstract
As a member of the signal transducer and activator of transcription (STAT) family, STAT3 plays a critical role in several biological pathways such as cell proliferation, migration, survival, and differentiation. Due to abnormal continuous activation in tumors, inhibition of STAT3 has emerged as an attractive approach for the treatment of various cancer cells. Herein, we report a series of novel STAT3 inhibitors based on benzo[b]thiophene 1,1-dioxide scaffold and evaluated their anticancer potency. Among them, compound 8b exhibited the best activity against cancer cells. Compound 8b induced apoptosis and blocked the cell cycle. Meanwhile, 8b reduced intracellular ROS content and caused the loss of mitochondrial membrane potential. Further research revealed that 8b significantly blocked STAT3 phosphorylation and STAT3-dependent dual-luciferase reporter gene experiments showed that compound 8b has a marked inhibition of STAT3-mediated Firefly luciferase activity. Molecular modeling studies revealed compound 8b occupied the pocket well with the SH2 domain in a favorable conformation.
Collapse
Affiliation(s)
- Wen-Zhen Li
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hui-Zhi Xi
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi-Jie Wang
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong-Bo Ma
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhi-Qiang Cheng
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yu Yang
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Meng-Ling Wu
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ting-Mei Liu
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wen Yang
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qin Wang
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Meng-Ya Liao
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Xia
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi-Wen Zhang
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Su C, Liu S, Ma X, Liu J, Liu J, Lei M, Cao Y. The effect and mechanism of erianin on the reversal of oxaliplatin resistance in human colon cancer cells. Cell Biol Int 2021; 45:2420-2428. [PMID: 34351659 DOI: 10.1002/cbin.11684] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/19/2021] [Accepted: 08/01/2021] [Indexed: 11/06/2022]
Abstract
Multidrug resistance (MDR) is the main cause of chemotherapy failure in the treatment of colon cancer and the high expression of drug efflux protein P-gp is one of the main factors of MDR. P-gp expression is regulated by the signal transducer and activator of transcription 3 (STAT3) signaling pathway. In this study, human colon cancer oxaliplatin-resistant cells were treated with oxaliplatin combined with the natural product erianin. Then, we evaluated the impact of erianin on drug resistance, and explored the relationship between erianin-related oxaliplatin resistance and the Janus kinase 2/STAT3 signaling pathway in vitro. Our research showed that erianin could significantly inhibit the proliferation of human colon cancer oxaliplatin-resistant cells, and suppress the cell cycle of oxaliplatin-resistant cells in the G2/M phase, indicating that erianin could regulate the MDR phenotype of oxaliplatin-resistant cells, and its mechanism might be the inhibition of STAT3 signaling pathway and the significant reduction of P-gp expression. However, this study provides a theoretical basis for the clinical application of erianin in platinum-based chemotherapy for colon cancer.
Collapse
Affiliation(s)
- Chang Su
- Department of Surgery, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Shaoqun Liu
- Department of Surgery, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Xiaoying Ma
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, PR China
| | - Jiajun Liu
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, PR China
| | - Jianwen Liu
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, PR China
| | - Ming Lei
- Department of Surgery, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Yiou Cao
- Department of Surgery, Minhang Hospital, Fudan University, Shanghai, PR China
| |
Collapse
|
17
|
Liu CL, Yuan RH, Mao TL. The Molecular Landscape Influencing Prognoses of Epithelial Ovarian Cancer. Biomolecules 2021; 11:998. [PMID: 34356623 PMCID: PMC8301761 DOI: 10.3390/biom11070998] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 12/26/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is one of the major increasing lethal malignancies of the gynecological tract, mostly due to delayed diagnosis and chemoresistance, as well as its very heterogeneous genetic makeup. Application of high-throughput molecular technologies, gene expression microarrays, and powerful preclinical models has provided a deeper understanding of the molecular characteristics of EOC. Therefore, molecular markers have become a potent tool in EOC management, including prediction of aggressiveness, prognosis, and recurrence, and identification of novel therapeutic targets. In addition, biomarkers derived from genomic/epigenomic alterations (e.g., gene mutations, copy number aberrations, and DNA methylation) enable targeted treatment of affected signaling pathways in advanced EOC, thereby improving the effectiveness of traditional treatments. This review outlines the molecular landscape and discusses the impacts of biomarkers on the detection, diagnosis, surveillance, and therapeutic targets of EOC. These findings focus on the necessity to translate these potential biomarkers into clinical practice.
Collapse
Affiliation(s)
- Chao-Lien Liu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- PhD Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Ray-Hwang Yuan
- Department of Surgery, National Taiwan University Hospital, Taipei 10002, Taiwan;
- Department of Surgery, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Tsui-Lien Mao
- Department of Pathology, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei 10002, Taiwan
| |
Collapse
|
18
|
A novel small molecule LLL12B inhibits STAT3 signaling and sensitizes ovarian cancer cell to paclitaxel and cisplatin. PLoS One 2021; 16:e0240145. [PMID: 33909625 PMCID: PMC8081214 DOI: 10.1371/journal.pone.0240145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 04/01/2021] [Indexed: 11/19/2022] Open
Abstract
Ovarian cancer is the fifth most common cause of cancer deaths among American women. Platinum and taxane combination chemotherapy represents the first-line approach for ovarian cancer, but treatment success is often limited by chemoresistance. Therefore, it is necessary to find new drugs to sensitize ovarian cancer cells to chemotherapy. Persistent activation of Signal Transducer and Activator of Transcription 3 (STAT3) signaling plays an important role in oncogenesis. Using a novel approach called advanced multiple ligand simultaneous docking (AMLSD), we developed a novel nonpeptide small molecule, LLL12B, which targets the STAT3 pathway. In this study, LLL12B inhibited STAT3 phosphorylation (tyrosine 705) and the expression of its downstream targets, which are associated with cancer cell proliferation and survival. We showed that LLL12B also inhibits cell viability, migration, and proliferation in human ovarian cancer cells. LLL12B combined with either paclitaxel or with cisplatin demonstrated synergistic inhibitory effects relative to monotherapy in inhibiting cell viability and LLL12B-paclitaxel or LLL12B-cisplatin combination exhibited greater inhibitory effects than cisplatin-paclitaxel combination in ovarian cancer cells. Furthermore, LLL12B-paclitaxel or LLL12B-cisplatin combination showed more significant in inhibiting cell migration and growth than monotherapy in ovarian cancer cells. In summary, our results support the novel small molecule LLL12B as a potent STAT3 inhibitor in human ovarian cancer cells and suggest that LLL12B in combination with the current front-line chemotherapeutic drugs cisplatin and paclitaxel may represent a promising approach for ovarian cancer therapy.
Collapse
|
19
|
STAT3 and p53: Dual Target for Cancer Therapy. Biomedicines 2020; 8:biomedicines8120637. [PMID: 33371351 PMCID: PMC7767392 DOI: 10.3390/biomedicines8120637] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/15/2020] [Accepted: 12/19/2020] [Indexed: 02/06/2023] Open
Abstract
The tumor suppressor p53 is considered the "guardian of the genome" that can protect cells against cancer by inducing cell cycle arrest followed by cell death. However, STAT3 is constitutively activated in several human cancers and plays crucial roles in promoting cancer cell proliferation and survival. Hence, STAT3 and p53 have opposing roles in cellular pathway regulation, as activation of STAT3 upregulates the survival pathway, whereas p53 triggers the apoptotic pathway. Constitutive activation of STAT3 and gain or loss of p53 function due to mutations are the most frequent events in numerous cancer types. Several studies have reported the association of STAT3 and/or p53 mutations with drug resistance in cancer treatment. This review discusses the relationship between STAT3 and p53 status in cancer, the molecular mechanism underlying the negative regulation of p53 by STAT3, and vice versa. Moreover, it underlines prospective therapies targeting both STAT3 and p53 to enhance chemotherapeutic outcomes.
Collapse
|
20
|
Garg M, Shanmugam MK, Bhardwaj V, Goel A, Gupta R, Sharma A, Baligar P, Kumar AP, Goh BC, Wang L, Sethi G. The pleiotropic role of transcription factor STAT3 in oncogenesis and its targeting through natural products for cancer prevention and therapy. Med Res Rev 2020; 41:1291-1336. [PMID: 33289118 DOI: 10.1002/med.21761] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/30/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is one of the crucial transcription factors, responsible for regulating cellular proliferation, cellular differentiation, migration, programmed cell death, inflammatory response, angiogenesis, and immune activation. In this review, we have discussed the classical regulation of STAT3 via diverse growth factors, cytokines, G-protein-coupled receptors, as well as toll-like receptors. We have also highlighted the potential role of noncoding RNAs in regulating STAT3 signaling. However, the deregulation of STAT3 signaling has been found to be associated with the initiation and progression of both solid and hematological malignancies. Additionally, hyperactivation of STAT3 signaling can maintain the cancer stem cell phenotype by modulating the tumor microenvironment, cellular metabolism, and immune responses to favor drug resistance and metastasis. Finally, we have also discussed several plausible ways to target oncogenic STAT3 signaling using various small molecules derived from natural products.
Collapse
Affiliation(s)
- Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vipul Bhardwaj
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Akul Goel
- La Canada High School, La Canada Flintridge, California, USA
| | - Rajat Gupta
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Arundhiti Sharma
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Prakash Baligar
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, Center for Translational Medicine, Singapore, Singapore
| | - Boon Cher Goh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, Center for Translational Medicine, Singapore, Singapore
- Department of Hematology-Oncology, National University Health System, Singapore, Singapore
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, Center for Translational Medicine, Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
21
|
Beyranvand Nejad E, Labrie C, van der Sluis TC, van Duikeren S, Franken KLMC, Roosenhoff R, Arens R, van Hall T, van der Burg SH. Interleukin-6-mediated resistance to immunotherapy is linked to impaired myeloid cell function. Int J Cancer 2020; 148:211-225. [PMID: 32875568 PMCID: PMC7693233 DOI: 10.1002/ijc.33280] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/30/2020] [Accepted: 08/14/2020] [Indexed: 12/17/2022]
Abstract
High serum levels of interleukin-6 (IL-6) correlate with poor prognosis and chemotherapy resistance in several cancers. The underlying mechanisms and its effects on immunotherapy are largely unknown. To address this, we developed a human papillomavirus type 16 (HPV16)-associated tumor model expressing IL-6 to investigate the impact of tumor-expressed IL-6 during cisplatin chemotherapy and HPV16 synthetic long peptide vaccination as immunotherapy. The effects of tumor-produced IL-6 on tumor growth, survival and the tumor microenvironment were analyzed. Our data demonstrated that tumor-produced IL-6 conferred resistance to cisplatin and therapeutic vaccination. This was not caused by a changed in vitro or in vivo growth rate of tumor cells, or a changed sensitivity of tumor cells to chemotherapy or T-cell-mediated killing. Furthermore, no overt differences in the frequencies of tumor-infiltrating subsets of T cells or CD11b+ myeloid cells were observed. IL-6, however, affected the systemic and local function of myeloid cells, reflected by a strong reduction of major histocompatibility complex (MHC) class II expression on all major myeloid cell subtypes. Resistance to both therapies was associated with a changed intratumoral influx of MHC class II+ myeloid cells toward myeloid cells with no or lower MHC class II expression. Importantly, while these IL-6-mediated effects provided resistance to the immunotherapy and chemotherapy as single therapies, their combination still successfully mediated tumor control. In conclusion, IL-6-mediated therapy resistance is caused by an extrinsic mechanism involving an impaired function of intratumoral myeloid cells. The fact that resistance can be overcome by combination therapies provides direction to more effective therapies for cancer.
Collapse
Affiliation(s)
- Elham Beyranvand Nejad
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Camilla Labrie
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Tetje C van der Sluis
- Department of Immunology, Leiden University Medical Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne van Duikeren
- Department of Immunology, Leiden University Medical Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Kees L M C Franken
- Department of Immunology, Leiden University Medical Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Rueshandra Roosenhoff
- Department of Immunology, Leiden University Medical Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Ramon Arens
- Department of Immunology, Leiden University Medical Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
22
|
Su Q, Banks E, Bebernitz G, Bell K, Borenstein CF, Chen H, Chuaqui CE, Deng N, Ferguson AD, Kawatkar S, Grimster NP, Ruston L, Lyne PD, Read JA, Peng X, Pei X, Fawell S, Tang Z, Throner S, Vasbinder MM, Wang H, Winter-Holt J, Woessner R, Wu A, Yang W, Zinda M, Kettle JG. Discovery of (2R)-N-[3-[2-[(3-Methoxy-1-methyl-pyrazol-4-yl)amino]pyrimidin-4-yl]-1H-indol-7-yl]-2-(4-methylpiperazin-1-yl)propenamide (AZD4205) as a Potent and Selective Janus Kinase 1 Inhibitor. J Med Chem 2020; 63:4517-4527. [DOI: 10.1021/acs.jmedchem.9b01392] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Qibin Su
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Erica Banks
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | | | - Kirsten Bell
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | | | - Huawei Chen
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Claudio E. Chuaqui
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Nanhua Deng
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Andrew D. Ferguson
- Discovery Sciences, R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Sameer Kawatkar
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Neil P. Grimster
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Linette Ruston
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Paul D. Lyne
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Jon A. Read
- Discovery Sciences, R&D, AstraZeneca R&D, Cambridge CB4 0WG, U.K
| | - Xianyou Peng
- Pharmaron Beijing Co., Ltd., 6 Taihe Road, BDA, Beijing 100176, China
| | - Xiaohui Pei
- Pharmaron Beijing Co., Ltd., 6 Taihe Road, BDA, Beijing 100176, China
| | - Stephen Fawell
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Zhanlei Tang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road, BDA, Beijing 100176, China
| | - Scott Throner
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | | | - Haoyu Wang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road, BDA, Beijing 100176, China
| | | | - Richard Woessner
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Allan Wu
- Discovery Sciences, R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | - Wenzhan Yang
- Early Product Development, Pharmaceutical Sciences, R&D, Boston, Massachusetts 02451, United States
| | - Michael Zinda
- Oncology R&D, AstraZeneca R&D, Waltham, Massachusetts 02451, United States
| | | |
Collapse
|
23
|
Letra‐Vilela R, Cardoso B, Silva‐Almeida C, Maia Rocha A, Murtinheira F, Branco‐Santos J, Rodriguez C, Martin V, Santa‐Marta M, Herrera F. Can asymmetric post-translational modifications regulate the behavior of STAT3 homodimers? FASEB Bioadv 2020; 2:116-125. [PMID: 32123861 PMCID: PMC7003655 DOI: 10.1096/fba.2019-00049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 06/14/2019] [Accepted: 12/10/2019] [Indexed: 01/16/2023] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a ubiquitous and pleiotropic transcription factor that plays essential roles in normal development, immunity, response to tissue damage and cancer. We have developed a Venus-STAT3 bimolecular fluorescence complementation assay that allows the visualization and study of STAT3 dimerization and protein-protein interactions in living cells. Inactivating mutations on residues susceptible to post-translational modifications (PTMs) (K49R, K140R, K685R, Y705F and S727A) changed significantly the intracellular distribution of unstimulated STAT3 dimers when the dimers were formed by STAT3 molecules that carried different mutations (ie they were "asymmetric"). Some of these asymmetric dimers changed the proliferation rate of HeLa cells. Our results indicate that asymmetric PTMs on STAT3 dimers could constitute a new level of regulation of STAT3 signaling. We put forward these observations as a working hypothesis, since confirming the existence of asymmetric STAT3 homodimers in nature is extremely difficult, and our own experimental setup has technical limitations that we discuss. However, if our hypothesis is confirmed, its conceptual implications go far beyond STAT3, and could advance our understanding and control of signaling pathways.
Collapse
Affiliation(s)
- Ricardo Letra‐Vilela
- Cell Structure and Dynamics LaboratoryInstituto de Tecnologia Quimica e Biologica (ITQB‐NOVA)Universidade Nova de LisboaOeirasPortugal
- Cell Structure and Dynamics LaboratoryFaculdade de CiênciasUniversidade de LisboaLisbonPortugal
| | - Beatriz Cardoso
- Cell Structure and Dynamics LaboratoryInstituto de Tecnologia Quimica e Biologica (ITQB‐NOVA)Universidade Nova de LisboaOeirasPortugal
| | - Catarina Silva‐Almeida
- Cell Structure and Dynamics LaboratoryInstituto de Tecnologia Quimica e Biologica (ITQB‐NOVA)Universidade Nova de LisboaOeirasPortugal
| | - Ana Maia Rocha
- Cell Structure and Dynamics LaboratoryInstituto de Tecnologia Quimica e Biologica (ITQB‐NOVA)Universidade Nova de LisboaOeirasPortugal
| | - Fernanda Murtinheira
- Cell Structure and Dynamics LaboratoryInstituto de Tecnologia Quimica e Biologica (ITQB‐NOVA)Universidade Nova de LisboaOeirasPortugal
- Cell Structure and Dynamics LaboratoryFaculdade de CiênciasUniversidade de LisboaLisbonPortugal
| | - Joana Branco‐Santos
- Cell Structure and Dynamics LaboratoryInstituto de Tecnologia Quimica e Biologica (ITQB‐NOVA)Universidade Nova de LisboaOeirasPortugal
| | - Carmen Rodriguez
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA) and Departamento de Morfología y Biología CelularFacultad de MedicinaUniversity of OviedoOviedoSpain
| | - Vanesa Martin
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA) and Departamento de Morfología y Biología CelularFacultad de MedicinaUniversity of OviedoOviedoSpain
| | - Mariana Santa‐Marta
- Cell Structure and Dynamics LaboratoryInstituto de Tecnologia Quimica e Biologica (ITQB‐NOVA)Universidade Nova de LisboaOeirasPortugal
| | - Federico Herrera
- Cell Structure and Dynamics LaboratoryInstituto de Tecnologia Quimica e Biologica (ITQB‐NOVA)Universidade Nova de LisboaOeirasPortugal
- Cell Structure and Dynamics LaboratoryFaculdade de CiênciasUniversidade de LisboaLisbonPortugal
| |
Collapse
|
24
|
Bagratuni T, Mavrianou N, Gavalas NG, Tzannis K, Arapinis C, Liontos M, Christodoulou MI, Thomakos N, Haidopoulos D, Rodolakis A, Kastritis E, Scorilas A, Dimopoulos MA, Bamias A. JQ1 inhibits tumour growth in combination with cisplatin and suppresses JAK/STAT signalling pathway in ovarian cancer. Eur J Cancer 2020; 126:125-135. [PMID: 31927213 DOI: 10.1016/j.ejca.2019.11.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/02/2019] [Accepted: 11/09/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Overexpression of c-Myc is commonly seen in human ovarian cancers, and this could be a potentially novel therapeutic target for this disease. JQ1, a selective small-molecule BET (Bromodomain and extraterminal domain family) bromodomain (BRDs) inhibitor, has been found to suppress tumour progression in several cancer cell types. RESULTS Using a panel of ovarian cancer cell lines and primary cell cultures from human ovarian cancer ascites, we demonstrated that JQ1 significantly suppressed cell proliferation and induced apoptosis in an ovarian cancer cell by targeting BRD4 and c-Μyc. In addition, JQ1 sensitized ovarian cancer cells to cisplatin, the most commonly used chemotherapeutic agent in ovarian cancer. Importantly, this effect was observed in ovarian cells, which exhibited resistance to cisplatin alone. Finally, we show that JQ1 interacts with the JAK-STAT signalling pathway, a pathway important in supporting ovarian cancer cell survival by suppressing or inducing genes involved in cell survival and apoptosis, respectively. CONCLUSION Our data, taken together, suggest that JQ1 is an attractive antitumour candidate for further investigation in the treatment of ovarian cancer, as it associates with cell proliferation, apoptosis, and alterations in the JAK-STAT signalling pathway, especially in patients with a platinum-resistant profile or in patients with relapsed disease.
Collapse
Affiliation(s)
- Tina Bagratuni
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Greece
| | - Nefeli Mavrianou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Greece
| | - Nikolaos G Gavalas
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Greece
| | - Kimon Tzannis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Greece
| | - Calliope Arapinis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Greece
| | - Michael Liontos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Greece
| | - Maria I Christodoulou
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Greece
| | - Nikolaos Thomakos
- 1(st) Department of Obstetrics & Gynecology, National and Kapodistrian University of Athens, 'Alexandra' Hospital, Athens, Greece
| | - Dimitrios Haidopoulos
- 1(st) Department of Obstetrics & Gynecology, National and Kapodistrian University of Athens, 'Alexandra' Hospital, Athens, Greece
| | - Alexandros Rodolakis
- 1(st) Department of Obstetrics & Gynecology, National and Kapodistrian University of Athens, 'Alexandra' Hospital, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Greece
| | - Aristotle Bamias
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
25
|
Liang R, Chen X, Chen L, Wan F, Chen K, Sun Y, Zhu X. STAT3 signaling in ovarian cancer: a potential therapeutic target. J Cancer 2020; 11:837-848. [PMID: 31949487 PMCID: PMC6959025 DOI: 10.7150/jca.35011] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 09/08/2019] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence has shown that Signal Transducer and Activator of Transcription 3 (STAT3) is thought to be a promising target for cancer therapy as STAT3 is frequently overexpressed in a wide range of cancer cells as well as clinical specimens, promoting tumor progression. It is widely accepted that STAT3 regulates a variety of cellular processes, such as tumor cell growth, survival, invasion, cancer stem cell-like characteristic, angiogenesis and drug-resistance. In this review, we focus on the role of STAT3 in tumorigenesis in ovarian cancer and discuss the existing inhibitors of STAT3 signaling that can be promisingly developed as the strategies for ovarian cancer therapy.
Collapse
Affiliation(s)
- Renba Liang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, P.R. China
| | - Xishan Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, P.R. China
| | - Li Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, P.R. China
| | - Fangzhu Wan
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, P.R. China
| | - Kaihua Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, P.R. China
| | - Yongchu Sun
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, P.R. China
| | - Xiaodong Zhu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, P.R. China
| |
Collapse
|
26
|
Wu CJ, Sundararajan V, Sheu BC, Huang RYJ, Wei LH. Activation of STAT3 and STAT5 Signaling in Epithelial Ovarian Cancer Progression: Mechanism and Therapeutic Opportunity. Cancers (Basel) 2019; 12:cancers12010024. [PMID: 31861720 PMCID: PMC7017004 DOI: 10.3390/cancers12010024] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal of all gynecologic malignancies. Despite advances in surgical and chemotherapeutic options, most patients with advanced EOC have a relapse within three years of diagnosis. Unfortunately, recurrent disease is generally not curable. Recent advances in maintenance therapy with anti-angiogenic agents or Poly ADP-ribose polymerase (PARP) inhibitors provided a substantial benefit concerning progression-free survival among certain women with advanced EOC. However, effective treatment options remain limited in most recurrent cases. Therefore, validated novel molecular therapeutic targets remain urgently needed in the management of EOC. Signal transducer and activator of transcription-3 (STAT3) and STAT5 are aberrantly activated through tyrosine phosphorylation in a wide variety of cancer types, including EOC. Extrinsic tumor microenvironmental factors in EOC, such as inflammatory cytokines, growth factors, hormones, and oxidative stress, can activate STAT3 and STAT5 through different mechanisms. Persistently activated STAT3 and, to some extent, STAT5 increase EOC tumor cell proliferation, survival, self-renewal, angiogenesis, metastasis, and chemoresistance while suppressing anti-tumor immunity. By doing so, the STAT3 and STAT5 activation in EOC controls properties of both tumor cells and their microenvironment, driving multiple distinct functions during EOC progression. Clinically, increasing evidence indicates that the activation of the STAT3/STAT5 pathway has significant correlation with reduced survival of recurrent EOC, suggesting the importance of STAT3/STAT5 as potential therapeutic targets for cancer therapy. This review summarizes the distinct role of STAT3 and STAT5 activities in the progression of EOC and discusses the emerging therapies specifically targeting STAT3 and STAT5 signaling in this disease setting.
Collapse
Affiliation(s)
- Chin-Jui Wu
- Department of Obstetrics & Gynecology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; (C.-J.W.); (B.-C.S.)
| | - Vignesh Sundararajan
- Cancer Science Institute of Singapore, National University of Singapore, Center for Translational Medicine, Singapore 117599, Singapore;
| | - Bor-Ching Sheu
- Department of Obstetrics & Gynecology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; (C.-J.W.); (B.-C.S.)
| | - Ruby Yun-Ju Huang
- Department of Obstetrics and Gynaecology, National University of Singapore, Singapore 119077, Singapore;
- School of Medicine, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Lin-Hung Wei
- Department of Obstetrics & Gynecology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; (C.-J.W.); (B.-C.S.)
- Correspondence: ; Tel.: +886-2-2312-3456 (ext. 71570); Fax: +886-2-2311-4965
| |
Collapse
|
27
|
Li H, Qian Y, Wang X, Pi R, Zhao X, Wei X. Targeted activation of Stat3 in combination with paclitaxel results in increased apoptosis in epithelial ovarian cancer cells and a reduced tumour burden. Cell Prolif 2019; 53:e12719. [PMID: 31778258 PMCID: PMC6985655 DOI: 10.1111/cpr.12719] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 02/05/2023] Open
Abstract
Objectives Stat3 is persistently activated in ovarian cancer cells, with a crucial role in tumour onset and progression. In this study, we examined the anti‐tumour effect of a small‐molecule inhibitor napabucasin (BBI608) on epithelial ovarian cancer (EOC) in vitro and in vivo, and investigated the underlying molecular mechanism of this drug in combination with paclitaxel. Materials and Methods A total of 156 ovarian cancer patient samples were analysed to determine the correlation between pStat3 expression in tumour cells and the prognosis of EOC patients. The anti‐tumour effect of BBI608 and/or paclitaxel on ovarian cancer in vitro was evaluated by CCK‐8, flow cytometry, Western blot and transwell assays. An in vivo intraperitoneal model was performed to confirm the effect of BBI608 on pStat3‐mediated peritoneal metastasis when combined with paclitaxel. Results Patients with high expression of pStat3 had poorer overall survival and progression‐free survival than those with low pStat3 expression. The synergy of BBI608 in combination with paclitaxel exerted dramatic growth inhibition and induced apoptosis in EOC cell lines. In vivo, the combination of two drugs significantly decreased intraperitoneal tumour burden and ascites volume, prolonged survival of tumour‐bearing mice compared with each monotherapy; these results were associated with downregulation of phospho‐Stat3 and activation of apoptosis pathway. Conclusions Targeting the activation of Stat3 may be a potential therapeutic approach for EOC by acting synergistically with paclitaxel.
Collapse
Affiliation(s)
- Hongyi Li
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Yanping Qian
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xi Wang
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Ruyu Pi
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| |
Collapse
|
28
|
Chong KY, Kang M, Garofalo F, Ueno D, Liang H, Cady S, Madarikan O, Pitruzzello N, Tsai CH, Hartwich TMP, Shuch BM, Yang-Hartwich Y. Inhibition of Heat Shock Protein 90 suppresses TWIST1 Transcription. Mol Pharmacol 2019; 96:168-179. [PMID: 31175180 DOI: 10.1124/mol.119.116137] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/03/2019] [Indexed: 02/14/2025] Open
Abstract
Molecular chaperone heat shock protein 90 (HSP90) is involved in oncogenic signaling pathways including epithelial-mesenchymal transition (EMT), a key process in tumor initiation, progression, metastasis, and chemoresistance. The molecular mechanisms underlying the involvement of HSP90 in EMT are still under investigation. In this study, we identified a previously unrecognized role of HSP90 in cooperating with signal transducer and activator of transcription 3 (STAT3) to regulate TWIST1 transcription in cancer cells. The HSP90 inhibitor 17-N-allylamino-17-demethoxygeldanamycin suppressed TWIST1 mRNA expression and promoter activity in epithelial ovarian cancer, renal clear cell cancer, and nasopharyngeal cancer cell lines. The interactions between HSP90 and transcription factors were visualized in cancer cell lines and tumor tissues using proximity ligation assays. Our findings reveal that HSP90 promotes the binding of STAT3 to the TWIST1 promoter, leading to the transcription of TWIST1. The inhibition of HSP90 downregulates STAT3 activity and TWIST1 transcription, thereby suppressing EMT and potentially inhibiting tumor progression, metastasis, and chemoresistance in different types of cancers. SIGNIFICANCE STATEMENT: Our study provides new evidence that HSP90 promotes EMT through enhancing TWIST1 transcription, which can be suppressed by HSP90 inhibitors. The HSP90 inhibitor inhibits EMT, thus potentially slowing down tumor growth, invasion, dissemination, metastasis, and drug resistance. These findings will hopefully pave the way for new therapeutic opportunities to target EMT and metastasis using HSP90 inhibitors.
Collapse
Affiliation(s)
- Kay Yi Chong
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut (K.Y.C., M.K., F.G., S.C., T.M.P.H., Y.Y.-H.); The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (M.K.); Ronald Reagan UCLA Medical Center, University of California Los Angeles, Santa Monica, California (D.U., B.M.S.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (H.L.); Department of Biology and Environmental Science, University of New Haven, West Haven, Connecticut (O.M., N.P., C.-H.T.); and Yale Cancer Center, New Haven, Connecticut (Y.Y.-H.)
| | - Min Kang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut (K.Y.C., M.K., F.G., S.C., T.M.P.H., Y.Y.-H.); The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (M.K.); Ronald Reagan UCLA Medical Center, University of California Los Angeles, Santa Monica, California (D.U., B.M.S.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (H.L.); Department of Biology and Environmental Science, University of New Haven, West Haven, Connecticut (O.M., N.P., C.-H.T.); and Yale Cancer Center, New Haven, Connecticut (Y.Y.-H.)
| | - Francesca Garofalo
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut (K.Y.C., M.K., F.G., S.C., T.M.P.H., Y.Y.-H.); The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (M.K.); Ronald Reagan UCLA Medical Center, University of California Los Angeles, Santa Monica, California (D.U., B.M.S.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (H.L.); Department of Biology and Environmental Science, University of New Haven, West Haven, Connecticut (O.M., N.P., C.-H.T.); and Yale Cancer Center, New Haven, Connecticut (Y.Y.-H.)
| | - Daiki Ueno
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut (K.Y.C., M.K., F.G., S.C., T.M.P.H., Y.Y.-H.); The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (M.K.); Ronald Reagan UCLA Medical Center, University of California Los Angeles, Santa Monica, California (D.U., B.M.S.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (H.L.); Department of Biology and Environmental Science, University of New Haven, West Haven, Connecticut (O.M., N.P., C.-H.T.); and Yale Cancer Center, New Haven, Connecticut (Y.Y.-H.)
| | - Huamao Liang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut (K.Y.C., M.K., F.G., S.C., T.M.P.H., Y.Y.-H.); The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (M.K.); Ronald Reagan UCLA Medical Center, University of California Los Angeles, Santa Monica, California (D.U., B.M.S.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (H.L.); Department of Biology and Environmental Science, University of New Haven, West Haven, Connecticut (O.M., N.P., C.-H.T.); and Yale Cancer Center, New Haven, Connecticut (Y.Y.-H.)
| | - Sarah Cady
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut (K.Y.C., M.K., F.G., S.C., T.M.P.H., Y.Y.-H.); The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (M.K.); Ronald Reagan UCLA Medical Center, University of California Los Angeles, Santa Monica, California (D.U., B.M.S.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (H.L.); Department of Biology and Environmental Science, University of New Haven, West Haven, Connecticut (O.M., N.P., C.-H.T.); and Yale Cancer Center, New Haven, Connecticut (Y.Y.-H.)
| | - Oluwagbemisola Madarikan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut (K.Y.C., M.K., F.G., S.C., T.M.P.H., Y.Y.-H.); The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (M.K.); Ronald Reagan UCLA Medical Center, University of California Los Angeles, Santa Monica, California (D.U., B.M.S.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (H.L.); Department of Biology and Environmental Science, University of New Haven, West Haven, Connecticut (O.M., N.P., C.-H.T.); and Yale Cancer Center, New Haven, Connecticut (Y.Y.-H.)
| | - Nicholas Pitruzzello
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut (K.Y.C., M.K., F.G., S.C., T.M.P.H., Y.Y.-H.); The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (M.K.); Ronald Reagan UCLA Medical Center, University of California Los Angeles, Santa Monica, California (D.U., B.M.S.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (H.L.); Department of Biology and Environmental Science, University of New Haven, West Haven, Connecticut (O.M., N.P., C.-H.T.); and Yale Cancer Center, New Haven, Connecticut (Y.Y.-H.)
| | - Cheng-Hsiu Tsai
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut (K.Y.C., M.K., F.G., S.C., T.M.P.H., Y.Y.-H.); The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (M.K.); Ronald Reagan UCLA Medical Center, University of California Los Angeles, Santa Monica, California (D.U., B.M.S.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (H.L.); Department of Biology and Environmental Science, University of New Haven, West Haven, Connecticut (O.M., N.P., C.-H.T.); and Yale Cancer Center, New Haven, Connecticut (Y.Y.-H.)
| | - Tobias M P Hartwich
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut (K.Y.C., M.K., F.G., S.C., T.M.P.H., Y.Y.-H.); The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (M.K.); Ronald Reagan UCLA Medical Center, University of California Los Angeles, Santa Monica, California (D.U., B.M.S.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (H.L.); Department of Biology and Environmental Science, University of New Haven, West Haven, Connecticut (O.M., N.P., C.-H.T.); and Yale Cancer Center, New Haven, Connecticut (Y.Y.-H.)
| | - Brian M Shuch
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut (K.Y.C., M.K., F.G., S.C., T.M.P.H., Y.Y.-H.); The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (M.K.); Ronald Reagan UCLA Medical Center, University of California Los Angeles, Santa Monica, California (D.U., B.M.S.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (H.L.); Department of Biology and Environmental Science, University of New Haven, West Haven, Connecticut (O.M., N.P., C.-H.T.); and Yale Cancer Center, New Haven, Connecticut (Y.Y.-H.)
| | - Yang Yang-Hartwich
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut (K.Y.C., M.K., F.G., S.C., T.M.P.H., Y.Y.-H.); The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (M.K.); Ronald Reagan UCLA Medical Center, University of California Los Angeles, Santa Monica, California (D.U., B.M.S.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (H.L.); Department of Biology and Environmental Science, University of New Haven, West Haven, Connecticut (O.M., N.P., C.-H.T.); and Yale Cancer Center, New Haven, Connecticut (Y.Y.-H.)
| |
Collapse
|
29
|
Lu T, Bankhead A, Ljungman M, Neamati N. Multi-omics profiling reveals key signaling pathways in ovarian cancer controlled by STAT3. Am J Cancer Res 2019; 9:5478-5496. [PMID: 31534498 PMCID: PMC6735387 DOI: 10.7150/thno.33444] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/18/2019] [Indexed: 12/21/2022] Open
Abstract
Inhibiting STAT3 signaling reduces tumor progression, metastasis and chemoresistance, however the precise molecular mechanism has not been fully delineated in ovarian cancer. Methods: In this study, we generated STAT3 knockout (KO) ovarian cancer cell lines. Effects of STAT3 KO on cell proliferation, migration and spheroid formation were assessed in vitro and effects on in vivo tumor growth were tested using several tumor xenograft models. We used multi-omic genome-wide profiling to identify multi-level (Bru-Seq, RNA-Seq, and MS Proteomic) expression signatures of STAT3 KO ovarian cancer cells. Results: We observed that deletion of STAT3 blocked cell proliferation and migration in vitro and suppressed tumor growth in mice. Deletion of STAT3 transcriptionally suppressed key genes involved in EMT, cell cycle progression, E2F signaling, and altered stemness markers. Notably, KO of STAT3 resulted in modulation of the expression of other STAT family members. Conclusion: Our study presents a rich, multi-faceted summary of the molecular mechanisms impacted by STAT3 deletion and provides new insight for STAT3's potential as a therapeutic target in ovarian cancer.
Collapse
|
30
|
Shoemaker RH, Fox JT, Juliana MM, Moeinpour FL, Grubbs CJ. Evaluation of the STAT3 inhibitor GLG‑302 for the prevention of estrogen receptor‑positive and ‑negative mammary cancers. Oncol Rep 2019; 42:1205-1213. [PMID: 31322250 DOI: 10.3892/or.2019.7219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/12/2019] [Indexed: 01/09/2023] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) plays a key role in the transformation of normal cells to cancerous cells. Although inhibitors of STAT3 have been shown to suppress the growth of multiple cancer types in vitro and in vivo, such agents are of particular interest for the prevention of breast cancer, which affects over 200,000 women and claims more than 40,000 lives in the United States each year. In the present study, we employed the MMTV/Neu transgenic mouse model, which develops estrogen receptor (ER)‑negative, Neu‑overexpressing tumors, and the Sprague‑Dawley (SD) rat model, which develops ER‑positive tumors upon exposure to the carcinogen 7,12‑dimethylbenz[a]anthracene (DMBA), to test the efficacy of the STAT3 inhibitor GLG‑302 in the prevention of mammary cancer. Orally administered GLG‑302 and its trizma salt derivative reduced mammary cancer incidence, multiplicity, and tumor weights in female MMTV/Neu mice, and GLG‑302 reduced tumor multiplicity and weights in female DMBA‑treated rats. Consistent with the mechanism of action of STAT3 inhibitors, the reductions in mammary tumors were correlated with decreases in STAT3 phosphorylation and cell proliferation. These data suggest that GLG‑302 is a novel agent with potential for prevention of mammary cancer and support the further development of STAT3 inhibitors for this cause.
Collapse
Affiliation(s)
- Robert H Shoemaker
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD 20850, USA
| | - Jennifer T Fox
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD 20850, USA
| | - Margaret M Juliana
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Fariba L Moeinpour
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Clinton J Grubbs
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
31
|
Gambi G, Di Simone E, Basso V, Ricci L, Wang R, Verma A, Elemento O, Ponzoni M, Inghirami G, Icardi L, Mondino A. The Transcriptional Regulator Sin3A Contributes to the Oncogenic Potential of STAT3. Cancer Res 2019; 79:3076-3087. [PMID: 30692217 DOI: 10.1158/0008-5472.can-18-0359] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/14/2018] [Accepted: 01/23/2019] [Indexed: 11/16/2022]
Abstract
Epigenetic silencing of promoter and enhancer regions is a common phenomenon in malignant cells. The transcription factor STAT3 is aberrantly activated in several tumors, where its constitutive acetylation accounts for the transcriptional repression of a number of tumor suppressor genes (TSG) via molecular mechanisms that remain to be understood. Using nucleophosmin-anaplastic lymphoma kinase-positive (NPM-ALK+) anaplastic large-cell lymphoma (ALCL) as model system, we found in cells and patient-derived tumor xenografts that STAT3 is constitutively acetylated as a result of ALK activity. STAT3 acetylation relied on intact ALK-induced PI3K- and mTORC1-dependent signaling and was sensitive to resveratrol. Resveratrol lowered STAT3 acetylation, rescued TSG expression, and induced ALCL apoptotic cell death. STAT3 constitutively bound the Sin3A transcriptional repressor complex, and both STAT3 and Sin3A bound the promoter region of silenced TSG via a resveratrol-sensitive mechanism. Silencing SIN3A caused reexpression of TSG, induced ALCL apoptotic cell death in vitro, and hindered ALCL tumorigenic potential in vivo. A constitutive STAT3-Sin3A interaction was also found in breast adenocarcinoma cells and proved critical for TSG silencing and cell survival. Collectively, these results suggest that oncogene-driven STAT3 acetylation and its constitutive association with Sin3A represent novel and concomitant events contributing to STAT3 oncogenic potential. SIGNIFICANCE: This study delineates the transcriptional regulatory complex Sin3A as a mediator of STAT3 transcriptional repressor activity and identifies the STAT3/Sin3A axis as a druggable target to antagonize STAT3-addicted tumors. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/79/12/3076/F1.large.jpg.See related commentary by Monteleone and Poli, p. 3031.
Collapse
Affiliation(s)
- Giovanni Gambi
- Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Di Simone
- Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Veronica Basso
- Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luisa Ricci
- Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rui Wang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
| | - Akanksha Verma
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York.,Institute for Precision Medicine, Weill Cornell Medical College, New York, New York.,Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York
| | - Olivier Elemento
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York.,Institute for Precision Medicine, Weill Cornell Medical College, New York, New York.,Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York
| | - Maurilio Ponzoni
- Department of Pathology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York.,Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies (CeRMS), University of Turin, Turin, Italy.,Department of Pathology and NYU Cancer Center, New York University School of Medicine, New York, New York
| | - Laura Icardi
- Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Anna Mondino
- Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
32
|
Vallée A, Guillevin R, Vallée JN. Vasculogenesis and angiogenesis initiation under normoxic conditions through Wnt/β-catenin pathway in gliomas. Rev Neurosci 2018; 29:71-91. [PMID: 28822229 DOI: 10.1515/revneuro-2017-0032] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 06/25/2017] [Indexed: 12/11/2022]
Abstract
The canonical Wnt/β-catenin pathway is up-regulated in gliomas and involved in proliferation, invasion, apoptosis, vasculogenesis and angiogenesis. Nuclear β-catenin accumulation correlates with malignancy. Hypoxia activates hypoxia-inducible factor (HIF)-1α by inhibiting HIF-1α prolyl hydroxylation, which promotes glycolytic energy metabolism, vasculogenesis and angiogenesis, whereas HIF-1α is degraded by the HIF prolyl hydroxylase under normoxic conditions. We focus this review on the links between the activated Wnt/β-catenin pathway and the mechanisms underlying vasculogenesis and angiogenesis through HIF-1α under normoxic conditions in gliomas. Wnt-induced epidermal growth factor receptor/phosphatidylinositol 3-kinase (PI3K)/Akt signaling, Wnt-induced signal transducers and activators of transcription 3 (STAT3) signaling, and Wnt/β-catenin target gene transduction (c-Myc) can activate HIF-1α in a hypoxia-independent manner. The PI3K/Akt/mammalian target of rapamycin pathway activates HIF-1α through eukaryotic translation initiation factor 4E-binding protein 1 and STAT3. The β-catenin/T-cell factor 4 complex directly binds to STAT3 and activates HIF-1α, which up-regulates the Wnt/β-catenin target genes cyclin D1 and c-Myc in a positive feedback loop. Phosphorylated STAT3 by interleukin-6 or leukemia inhibitory factor activates HIF-1α even under normoxic conditions. The activation of the Wnt/β-catenin pathway induces, via the Wnt target genes c-Myc and cyclin D1 or via HIF-1α, gene transactivation encoding aerobic glycolysis enzymes, such as glucose transporter, hexokinase 2, pyruvate kinase M2, pyruvate dehydrogenase kinase 1 and lactate dehydrogenase-A, leading to lactate production, as the primary alternative of ATP, at all oxygen levels, even in normoxic conditions. Lactate released by glioma cells via the monocarboxylate lactate transporter-1 up-regulated by HIF-1α and lactate anion activates HIF-1α in normoxic endothelial cells by inhibiting HIF-1α prolyl hydroxylation and preventing HIF labeling by the von Hippel-Lindau protein. Increased lactate with acid environment and HIF-1α overexpression induce the vascular endothelial growth factor (VEGF) pathway of vasculogenesis and angiogenesis under normoxic conditions. Hypoxia and acidic pH have no synergistic effect on VEGF transcription.
Collapse
Affiliation(s)
- Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, 11 Boulevard Marie et Pierre Curie, F-86000 Poitiers, France
| | - Rémy Guillevin
- DACTIM, UMR CNRS 7348, Université de Poitiers et CHU de Poitiers, F-86000 Poitiers, France
| | - Jean-Noël Vallée
- Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, University of Poitiers, F-86000 Poitiers, France
| |
Collapse
|
33
|
Grimster NP, Anderson E, Alimzhanov M, Bebernitz G, Bell K, Chuaqui C, Deegan T, Ferguson AD, Gero T, Harsch A, Huszar D, Kawatkar A, Kettle JG, Lyne P, Read JA, Rivard Costa C, Ruston L, Schroeder P, Shi J, Su Q, Throner S, Toader D, Vasbinder M, Woessner R, Wang H, Wu A, Ye M, Zheng W, Zinda M. Discovery and Optimization of a Novel Series of Highly Selective JAK1 Kinase Inhibitors. J Med Chem 2018; 61:5235-5244. [DOI: 10.1021/acs.jmedchem.8b00076] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Linette Ruston
- Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, Macclesfield SK10 2NA, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sasidharan Nair V, Toor SM, Ali BR, Elkord E. Dual inhibition of STAT1 and STAT3 activation downregulates expression of PD-L1 in human breast cancer cells. Expert Opin Ther Targets 2018; 22:547-557. [PMID: 29702007 DOI: 10.1080/14728222.2018.1471137] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Breast cancer is the most commonly diagnosed cancer, and it is a leading cause of cancer-related deaths in females worldwide. Triple-negative breast cancer (TNBC) constitutes 15% of breast cancer and shows distinct metastasis profiles with poor prognosis. Strong PD-L1 expression has been observed in some tumors, supporting their escape from immune surveillance. Targeting PD-L1 could be a promising therapeutic approach in breast cancer patients. We investigated potential molecular mechanisms for constitutive expression of PD-L1 by inhibiting upstream STAT1 and STAT3 signals. METHODS PD-L1 expression in three breast cancer cell lines was measured using quantitative PCR and western blotting. Activation of STAT1 and STAT3 was blocked using pharmacological inhibitors and siRNA. The mechanism underlying the constitutive expression of PD-L1 was investigated using ChIP and co-immunoprecipitation assays. RESULTS We found that individual inhibition of STAT1 and STAT3 activation partially downregulated PD-L1, while combined inhibition completely downregulated PD-L1 expression. Moreover, our results suggest that pSTAT1-pSTAT3 dimerize in cytosol and translocate to the nucleus, where they bind to PD-L1 promoter and induce PD-L1 expression. CONCLUSION These findings provide a rationale for combined targeting of STAT1 and STAT3 for the development of immune-based cancer therapies for down regulation of PD-L1 expression.
Collapse
Affiliation(s)
- Varun Sasidharan Nair
- a Cancer Research Center , Qatar Biomedical Research Institute, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation , Doha , Qatar
| | - Salman M Toor
- a Cancer Research Center , Qatar Biomedical Research Institute, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation , Doha , Qatar
| | - Bassam R Ali
- b College of Medicine and Health Sciences , United Arab Emirates University , Al Ain , United Arab Emirates
| | - Eyad Elkord
- a Cancer Research Center , Qatar Biomedical Research Institute, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation , Doha , Qatar.,c Institute of Cancer Sciences , University of Manchester , Manchester , UK
| |
Collapse
|
35
|
Liu SS, Liu N, Liu MY, Sun L, Xia WY, Lu HM, Fu YJ, Yang GL, Bo JJ, Liu XX, Feng H, Wu H, Li LF, Gao JX. An unusual intragenic promoter of PIWIL2 contributes to aberrant activation of oncogenic PL2L60. Oncotarget 2018; 8:46104-46120. [PMID: 28545024 PMCID: PMC5542253 DOI: 10.18632/oncotarget.17553] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 03/28/2017] [Indexed: 12/24/2022] Open
Abstract
PIWIL2-like (PL2L) protein 60 (PL2L60), a product of aberrantly activated PIWIL2 gene, is widely expressed in various types of tumors and may promote tumorigenesis. However, the mechanisms underlying the activation of expression of PL2L60 remain unknown. In this study, an intragenic promoter responsible for the activation of PL2L60 within the human PIWIL2 gene has been identified, cloned and characterized. The promoter of PL2L60 is located in the intron 10 of the host gene PIWIL2. Bioinformatic and mutagenic analysis reveals that this intragenic promoter within the sequence of 50 nucleotides contains two closely arranged cis-acting elements specific for the hepatic leukemia factor (HLF) in the positive strand and signal transducer and activator of transcription 3 (STAT3) in the negative strand. Chromatin immunoprecipitation analysis demonstrates that both the HLF and polymerase II (Pol II), a hallmark of active promoters, directly bind to the sequence, although STAT3 does not. Knockdown of HLF and STAT3 alone or both by RNA interference significantly reduced both promoter activity and the PL2L60 protein expression, although there is no additive effect. The expression of PL2L60 proteins was enhanced when host gene Piwil2 was genetically disrupted in a murine cell model. Taken together, we have identified a PL2L60-specific intragenic promoter in the host gene of PIWIL2, which is interdependently activated by HLF and STAT3 through steric interaction. This activation is dependent on cellular milieu rather than the integrity of host gene PIWIL2, highlighting a novel, important mechanism for a cancer-causing gene to be activated during tumorigenesis.
Collapse
Affiliation(s)
- Shan-Shan Liu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ning Liu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Meng-Yao Liu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Sun
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wu-Yan Xia
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hong-Min Lu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Jie Fu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Liang Yang
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Juan-Jie Bo
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Xing Liu
- Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haizhong Feng
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hailong Wu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lin-Feng Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Xin Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
36
|
PUM1 promotes ovarian cancer proliferation, migration and invasion. Biochem Biophys Res Commun 2018; 497:313-318. [DOI: 10.1016/j.bbrc.2018.02.078] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 02/07/2018] [Indexed: 01/30/2023]
|
37
|
Medhat AM, Azab KS, Said MM, El Fatih NM, El Bakary NM. Antitumor and radiosensitizing synergistic effects of apigenin and cryptotanshinone against solid Ehrlich carcinoma in female mice. Tumour Biol 2017; 39:1010428317728480. [PMID: 29022496 DOI: 10.1177/1010428317728480] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Considerable attention has been paid to the introduction of novel naturally occurring plant-derived radiosensitizer compounds in order to augment the radiation efficacy and improve the treatment outcome of different tumors. This study was therefore undertaken to evaluate the antitumor, antiangiogeneic, and synergistic radiosensitizing effects of apigenin, a dietary flavonoid, and/or cryptotanshinone, a terpenoid isolated from the roots of Salvia miltiorrhiza, against the growth of solid Ehrlich carcinoma in female mice. Apigenin (50 mg/kg body weight) and/or cryptotanshinone (40 mg/kg body weight) was intraperitoneally (i.p.) injected into non-irradiated or γ-irradiated (6.5 Gy whole-body γ-irradiation) solid Ehrlich carcinoma-bearing mice for 30 consecutive days. Investigations included molecular targets involved in proliferation, inflammation, angiogenesis, and tumor invasiveness. Treatment with apigenin and/or cryptotanshinone significantly suppressed the growth of solid Ehrlich carcinoma tumors and demonstrated a synergistic radiosensitizing efficacy together with γ-irradiation. These effects were achieved through downregulating the expression of angiogenic and lymphangiogenic regulators, including signal transducer and activator of transcription 3, vascular endothelial growth factor C, and tumor necrosis factor alpha, suppressing matrix metalloproteinase-2 and -9 activities, which play a key role in tumor invasion and metastasis, and enhancing apoptosis via inducing cleaved caspase-3 and granzyme B levels. Histological findings of solid Ehrlich carcinoma tumors verified the recorded data. In conclusion, a synergistic radiosensitizing efficacy for apigenin and cryptotanshinone was demonstrated against Ehrlich carcinoma in the current in vivo murine model, representing therefore a potential therapeutic strategy for increasing the radiation response of solid tumors.
Collapse
Affiliation(s)
- Amina M Medhat
- 1 Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Khaled Sh Azab
- 2 Radiation Biology Department, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Mahmoud M Said
- 1 Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Neama M El Fatih
- 2 Radiation Biology Department, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Nermeen M El Bakary
- 2 Radiation Biology Department, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
38
|
The prognostic values of signal transducers activators of transcription family in ovarian cancer. Biosci Rep 2017; 37:BSR20170650. [PMID: 28536310 PMCID: PMC5518537 DOI: 10.1042/bsr20170650] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/19/2017] [Accepted: 05/22/2017] [Indexed: 11/25/2022] Open
Abstract
Signal transducer and activator of transcription (STAT), a family of latent cytoplasmic transcription factors, are composed of seven identified members (STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b, STAT6). STATs are associated with several biological processes such as cell proliferation, invasion, and metastasis in various cancer types. In addition, the STAT family has been well studied as a prognostic predictor for a considerable number of solid tumors. However, the prognostic value of the STAT family in ovarian cancer patients remains unclear. In our present study, we intend to access the prognostic roles of the STAT family in ovarian carcinoma through the ‘Kaplan–Meier plotter’ (KM plotter) online database, which collected gene expression data and survival information (overall survival (OS)) from a total of 1582 ovarian cancer patients. Our results show that high mRNA expression of STAT1, STAT4, STAT5a, STAT5b, and STAT6, are correlated to a better OS of ovarian cancer patients, especially the high level of STAT1 and STAT4 are significantly related to a favorable OS for serous ovarian cancer patients. We further accessed the prognostic roles of individual STATs in other clinicopathological features, such as pathological grades, clinical stages, and TP53 mutation, and found that these genes indicate a favorable prognosis especially for late stage, poor differentiation, and TP53 mutated ovarian cancer patients. In conclusion, these results suggest that the STAT family plays a significant prognostic role in ovarian carcinoma and individual STATs, except STAT2 and STAT3, may act as favorable prognostic markers in ovarian cancer.
Collapse
|
39
|
Kim DH, Park JE, Chae IG, Park G, Lee S, Chun KS. Isoliquiritigenin inhibits the proliferation of human renal carcinoma Caki cells through the ROS-mediated regulation of the Jak2/STAT3 pathway. Oncol Rep 2017; 38:575-583. [PMID: 28560439 DOI: 10.3892/or.2017.5677] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 04/24/2017] [Indexed: 11/06/2022] Open
Abstract
Isoliquiritigenin (ISL) is a flavonoid with chalcone structure that has been noted in licorice and shallot, which are generally used in traditional Chinese medicine. ISL has demonstrated various pharmacological effects including antioxidant, anti-inflammatory and antitumor activity. However, the molecular mechanisms underlying the anticancer effects of ISL remain poorly understood. The present study revealed that ISL significantly decreased viability and induced apoptosis in human renal carcinoma Caki cells. The ISL-induced apoptosis was associated with the cleavage of caspase-9, -7 and -3, and that of PARP. Moreover, ISL increased the expression of pro-apoptotic protein Bax and diminished the expression of anti-apoptotic protein Bcl-2, and Bcl-xl, thereby increasing cytochrome c release. Treatment of cells with ISL also induced the expression of p53 through downregulation of murine double minute 2 (Mdm2). Furthermore, ISL generated reactive oxygen species (ROS), and pretreatment with ROS scavenger N-acetyl cysteine (NAC) and NADPH oxidase inhibitor diphenyleneiodonium abrogated the ISL-induced apoptosis. One of the key oncogenic signaling pathways is mediated through signal transducer and activator of transcription 3 (STAT3), which promotes abnormal cell proliferation. Incubation of cells with ISL markedly diminished phosphorylation and DNA binding activity of STAT3, and reduced expression of STAT3 responsive gene products, such as cyclin D1 and D2. ISL also attenuated constitutive phosphorylation of upstream kinase, Janus-activated kinase 2 (Jak2). Pretreatment with NAC abrogated the inhibitory effect of ISL on activation of STAT3 and blocked the cleavage of caspase-9, -7 and -3, and that of PARP in Caki cells. Taken together, the present study provides the first report that ISL induces apoptosis in Caki cells via generation of ROS, which causes induction of p53 and inhibition of the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Do-Hee Kim
- College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Ji Eun Park
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| | - In Gyeong Chae
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| | - Geumi Park
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| |
Collapse
|
40
|
Mahony R, Gargan S, Roberts KL, Bourke N, Keating SE, Bowie AG, O'Farrelly C, Stevenson NJ. A novel anti-viral role for STAT3 in IFN-α signalling responses. Cell Mol Life Sci 2017; 74:1755-1764. [PMID: 27988795 PMCID: PMC11107673 DOI: 10.1007/s00018-016-2435-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 11/16/2016] [Accepted: 12/05/2016] [Indexed: 10/20/2022]
Abstract
The cytokine, Interferon (IFN)-α, induces a wide spectrum of anti-viral mediators, via the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway. STAT1 and STAT2 are well characterised to upregulate IFN-stimulated gene (ISG) expression; but even though STAT3 is also activated by IFN-α, its role in anti-viral ISG induction is unclear. Several viruses, including Hepatitis C and Mumps, reduce cellular STAT3 protein levels, via the promotion of ubiquitin-mediated proteasomal degradation. This viral immune evasion mechanism suggests an undiscovered anti-viral role for STAT3 in IFN-α signalling. To investigate STAT3's functional involvement in this Type I IFN pathway, we first analysed its effect upon the replication of two viruses, Influenza and Vaccinia. Viral plaque assays, using Wild Type (WT) and STAT3-/- Murine Embryonic Fibroblasts (MEFs), revealed that STAT3 is required for the inhibition of Influenza and Vaccinia replication. Furthermore, STAT3 shRNA knockdown also enhanced Influenza replication and hindered induction of several, well characterised, anti-viral ISGs: PKR, OAS2, MxB and ISG15; while STAT3 expression had no effect upon induction of a separate ISG group: Viperin, IFI27, CXCL10 and CCL5. These discoveries reveal, for the first time, an anti-viral role for STAT3 in the IFN-α pathway and characterise a requirement for STAT3 in the expression of specific ISGs. These findings also identify STAT3 as a therapeutic target against viral infection and highlight it as an essential pathway component for endogenous and therapeutic IFN-α responsiveness.
Collapse
Affiliation(s)
- Rebecca Mahony
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Siobhán Gargan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Kim L Roberts
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Nollaig Bourke
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Sinead E Keating
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Nigel J Stevenson
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
41
|
Gao J, Chen J, Cai M, Xu H, Jiang J, Tong T, Wang H. Clustered localization of STAT3 during the cell cycle detected by super-resolution fluorescence microscopy. Methods Appl Fluoresc 2017; 5:024004. [DOI: 10.1088/2050-6120/aa6ab5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
42
|
Zheng J, Son DJ, Lee HL, Lee HP, Kim TH, Joo JH, Ham YW, Kim WJ, Jung JK, Han SB, Hong JT. (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol suppresses ovarian cancer cell growth via inhibition of ERK and STAT3. Mol Carcinog 2017; 56:2003-2013. [PMID: 28277616 DOI: 10.1002/mc.22648] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 02/01/2017] [Accepted: 03/03/2017] [Indexed: 12/14/2022]
Abstract
In the present study, we synthesized several non-aldehyde analogues of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal which showed anti-cancer effect. Interestingly, among the 16 compounds, we found that (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol (MMPP) showed the most significant anti-proliferative effect on PA-1 and SK-OV-3 ovarian epithelial cancer cells. MMPP treatment (0-15 µg/mL) induced apoptotic cell death, enhanced the expression of cleaved caspase-3, and cleaved caspase-9 in a concentration dependent manner. Notably, DNA binding activity of STAT3, phosphorylation of extracellular signal-regulated kinase (ERK) and p38 was significantly decreased by MMPP treatment. However, ERK siRNA augmented MMPP-induced inhibitory effect on cell growth rather than p38 siRNA or JNK siRNA. Moreover, combination treatment of MMPP with ERK inhibitor U0126 (10 µM) augmented MMPP-induced inhibitory effect on cell growth and DNA binding activity of STAT3, and enhanced expression of cleaved caspase-3 and cleaved caspase-9. In addition, STAT3 siRNA transfection augmented MMPP-induced cell growth inhibition. In PA-1 bearing xenograft mice model, MMPP (5 mg/kg) suppressed tumor growth significantly. Immunohistochemistry staining showed that the expression levels of p-ERK, PCNA, p-STAT3 were decreased while the expression level of caspase-3 was increased by MMPP treatment. Thus, MMPP may be a promising anti-cancer agent in ovarian epithelial cancer treatment.
Collapse
Affiliation(s)
- Jie Zheng
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hye Lim Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hee Pom Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Tae Hoon Kim
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jung Heun Joo
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Young Wan Ham
- Department of Chemistry, Utah Valley University 800 W, University Pkwy, Orem, Utah
| | - Wun Jae Kim
- College of Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jae Kyung Jung
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
43
|
Hilliard TS, Miklossy G, Chock C, Yue P, Williams P, Turkson J. 15α-methoxypuupehenol Induces Antitumor Effects In Vitro and In Vivo against Human Glioblastoma and Breast Cancer Models. Mol Cancer Ther 2017; 16:601-613. [PMID: 28069875 DOI: 10.1158/1535-7163.mct-16-0291] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 12/25/2016] [Accepted: 12/28/2016] [Indexed: 01/10/2023]
Abstract
Studies with 15α-methoxypuupehenol (15α-MP), obtained from the extracts of Hyrtios species, identified putative targets that are associated with its antitumor effects against human glioblastoma and breast cancer. In the human glioblastoma (U251MG) or breast cancer (MDA-MB-231) cells, treatment with 15α-MP repressed pY705Stat3, pErk1/2, pS147CyclinB1, pY507Alk (anaplastic lymphoma kinase), and pY478ezrin levels and induced pS10merlin, without inhibiting pJAK2 (Janus kinase) or pAkt induction. 15α-MP treatment induced loss of viability of breast cancer (MDA-MB-231, MDA-MB-468) and glioblastoma (U251MG) lines and glioblastoma patient-derived xenograft cells (G22) that harbor aberrantly active Stat3, with only moderate or little effect on the human breast cancer, MCF7, colorectal adenocarcinoma Caco-2, normal human lung fibroblast, WI-38, or normal mouse embryonic fibroblast (MEF Stat3fl/fl) lines that do not harbor constitutively active Stat3 or the Stat3-null (Stat3-/-) mouse astrocytes. 15α-MP-treated U251MG cells have severely impaired F-actin organization and altered morphology, including the cells rounding up, and undergo apoptosis, compared with a moderate, reversible morphology change observed for similarly treated mouse astrocytes. Treatment further inhibited U251MG or MDA-MB-231 cell proliferation, anchorage-independent growth, colony formation, and migration in vitro while only moderately or weakly affecting MCF7 cells or normal mouse astrocytes. Oral gavage delivery of 15α-MP inhibited the growth of U251MG subcutaneous tumor xenografts in mice, associated with apoptosis in the treated tumor tissues. Results together suggest that the modulation of Stat3, CyclinB1, Alk, ezrin, merlin, and Erk1/2 functions contributes to the antitumor effects of 15α-MP against glioblastoma and breast cancer progression. Mol Cancer Ther; 16(4); 601-13. ©2017 AACR.
Collapse
Affiliation(s)
- Tyvette S Hilliard
- Cancer Biology and Natural Products Program, University of Hawai'i Cancer Center, Honolulu, Hawaii
| | - Gabriella Miklossy
- Cancer Biology and Natural Products Program, University of Hawai'i Cancer Center, Honolulu, Hawaii
| | - Christopher Chock
- Department of Chemistry, University of Hawai'i at Manoa, Honolulu, Hawaii
| | - Peibin Yue
- Cancer Biology and Natural Products Program, University of Hawai'i Cancer Center, Honolulu, Hawaii
| | - Philip Williams
- Department of Chemistry, University of Hawai'i at Manoa, Honolulu, Hawaii
| | - James Turkson
- Cancer Biology and Natural Products Program, University of Hawai'i Cancer Center, Honolulu, Hawaii.
| |
Collapse
|
44
|
Natural product pectolinarigenin inhibits osteosarcoma growth and metastasis via SHP-1-mediated STAT3 signaling inhibition. Cell Death Dis 2016; 7:e2421. [PMID: 27735939 PMCID: PMC5133974 DOI: 10.1038/cddis.2016.305] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/24/2016] [Accepted: 08/26/2016] [Indexed: 12/22/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) has important roles in cancer aggressiveness and has been confirmed as an attractive target for cancer therapy. In this study, we used a dual-luciferase assay to identify that pectolinarigenin inhibited STAT3 activity. Further studies showed pectolinarigenin inhibited constitutive and interleukin-6-induced STAT3 signaling, diminished the accumulation of STAT3 in the nucleus and blocked STAT3 DNA-binding activity in osteosarcoma cells. Mechanism investigations indicated that pectolinarigenin disturbed the STAT3/DNA methyltransferase 1/HDAC1 histone deacetylase 1 complex formation in the promoter region of SHP-1, which reversely mediates STAT3 signaling, leading to the upregulation of SHP-1 expression in osteosarcoma. We also found pectolinarigenin significantly suppressed osteosarcoma cell proliferation, induced apoptosis and reduced the level of STAT3 downstream proteins cyclin D1, Survivin, B-cell lymphoma 2 (Bcl-2), B-cell lymphoma extra-large (Bcl-xl) and myeloid cell leukemia 1 (Mcl-1). In addition, pectolinarigenin inhibited migration, invasion and reserved epithelial–mesenchymal transition (EMT) phenotype in osteosarcoma cells. In spontaneous and patient-derived xenograft models of osteosarcoma, we identified administration (intraperitoneal) of pectolinarigenin (20 mg/kg/2 days and 50 mg/kg/2 days) blocked STAT3 activation and impaired tumor growth and metastasis with superior pharmacodynamic properties. Taken together, our findings demonstrate that pectolinarigenin may be a candidate for osteosarcoma intervention linked to its STAT3 signaling inhibitory activity.
Collapse
|
45
|
Ding DC, Liu HW, Chu TY. Interleukin-6 from Ovarian Mesenchymal Stem Cells Promotes Proliferation, Sphere and Colony Formation and Tumorigenesis of an Ovarian Cancer Cell Line SKOV3. J Cancer 2016; 7:1815-1823. [PMID: 27698921 PMCID: PMC5039365 DOI: 10.7150/jca.16116] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 07/18/2016] [Indexed: 12/19/2022] Open
Abstract
The origin of the majority of epithelial ovarian cancers (EOC) is regarded as extraovarian, with the ovary being the secondary site. The aim of this study was to explore the possible role of ovarian mesenchymal stem cells (OvMSCs) and secreted IL-6 in the development of EOC. OvMSCs were derived from normal ovarian stroma. Cell surface markers and differentiation capability were determined. The effects of IL-6 and conditioned medium of OvMSCs on the malignant phenotype of SKOV3 ovarian cancer cells were tested, and the status of STAT3 and ERK phosphorylation was investigated. OvMSCs had similar surface marker profiles as bone marrow mesenchymal stem cells, i.e., CD44 (+), CD90 (+) and CD45 (-), and was readily inducible to osteogenic, adipogenic and chondrogenic differentiation. OvMSCs secreted an extremely high level (>2500 pg/ml) of IL-6. Treatment of SKOV3 cells with conditioned media from OvMSCs increased cell proliferation, tumor sphere formation and anchorage independent growth, and resulted in activation of STAT3 but not ERK. Coinjection of OvMSCs with SKOV3 cell enhanced tumorigenesis in NOD-SCID mice. All of these behaviors were blocked by IL-6 receptor blocking antibody administered in vitro or in vivo. The OvMSCs alone injected into mice had no tumor growth after 3 months. By secreting high levels of IL-6, OvMSCs enhance the proliferation, sphere and colony formation and tumorigenesis of SKOV3 cells.
Collapse
Affiliation(s)
- Dah-Ching Ding
- Department of Obstetrics and Gynecology, Buddhist Tzu Chi General Hospital;; Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Hwan-Wun Liu
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan;; Department of Occupational Medicine, Buddhist Tzu Chi General Hospital
| | - Tang-Yuan Chu
- Department of Obstetrics and Gynecology, Buddhist Tzu Chi General Hospital;; Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan;; Cervical cancer prevention center, Buddhist Tzu Chi General Hospital
| |
Collapse
|
46
|
Khoogar R, Kim BC, Morris J, Wargovich MJ. Chemoprevention in gastrointestinal physiology and disease. Targeting the progression of cancer with natural products: a focus on gastrointestinal cancer. Am J Physiol Gastrointest Liver Physiol 2016; 310:G629-44. [PMID: 26893159 PMCID: PMC4867331 DOI: 10.1152/ajpgi.00201.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 02/11/2016] [Indexed: 01/31/2023]
Abstract
The last decade has witnessed remarkable progress in the utilization of natural products for the prevention and treatment of human cancer. Many agents now in the pipeline for clinical trial testing have evolved from our understanding of how human nutritional patterns account for widespread differences in cancer risk. In this review, we have focused on many of these promising agents arguing that they may provide a new strategy for cancer control: natural products once thought to be only preventive in their mode of action now are being explored for efficacy in tandem with cancer therapeutics. Natural products may reduce off-target toxicity of therapeutics while making cancers more amenable to therapy. On the horizon is the use of certain natural products, in their own right, as mitigants of late-stage cancer, a new frontier for small-molecule natural product drug discovery.
Collapse
Affiliation(s)
- Roxane Khoogar
- 1Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; and
| | - Byung-Chang Kim
- 2Center for Colorectal Center, Center for Cancer Prevention and Detection, Research Institute and Hospital, National Cancer Center, Ilsan-ro, Illsandong-gu, Goyang-si Gyeonggi-do, Republic of Korea
| | - Jay Morris
- 1Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; and
| | - Michael J. Wargovich
- 1Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; and
| |
Collapse
|
47
|
Hajimoradi M, Mohammad Hassan Z, Ebrahimi M, Soleimani M, Bakhshi M, Firouzi J, Samani FS. STAT3 is Overactivated in Gastric Cancer Stem-Like Cells. CELL JOURNAL 2016; 17:617-28. [PMID: 26862521 PMCID: PMC4746412 DOI: 10.22074/cellj.2016.3834] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 06/09/2015] [Indexed: 12/19/2022]
Abstract
Objective Gastric cancer (GC) is widely associated with chronic inflammation. The
pro inflammatory microenvironment provides conditions that disrupt stem/progenitor
cell proliferation and differentiation. The signal transducer and activator of transcrip-
tion-3 (STAT3) signaling pathway is involved in inflammation and also contributes to
the maintenance of embryonic stem cell (ESCs) pluripotency. Here, we have investi-
gated the activation status of STAT3 in GC stem-like cells (GCSLCs).
Materials and Methods In this experimental research, CSLCs derived from the human
GC cell line MKN-45 and patient specimens, through spheroid body formation, character-
ized and then assayed for the STAT3 transcription factor expression in mRNA and protein
level further to its activation.
Results Spheroid cells showed higher potential for spheroid formation than the pa-
rental cells. Furthemore, stemness genes NANOG, c-MYC and SOX-2 were over
expressed in spheroids of MKN-45 and in patient samples. In MKN-45 spheroid cells,
epithelial mesenchymal transition (EMT) related markers CDH2, SNAIL2, TWIST and
VIMENTIN were upregulated (P<0.05), but we observed no change in expression of
the E-cadherin epithelial marker. These cells exhibited more resistance to docetaxel
(DTX) when compared with parental cells (P<0.05) according to the MTS assay. Al-
though immunostaining and Western blotting showed expression of the STAT3 pro-
tein in both spheroids and parents, the mRNA level of STAT3 in spheroids was higher
than the parents. Nuclear translocation of STAT3 was accompanied by more intensive
phospho-STAT3 (p-STAT3) in spheroid structures relative to the parent cells accord-
ing to flow cytometry analysis (P<0.05).
Conclusion The present findings point to STAT3 over activation in GCSLCs. Com-
plementary experiments are required to extend the role of STAT3 in stemness fea-
tures and invasion properties of GCSCs and to consider the STAT3 pathway for CSC
targeted therapy.
Collapse
Affiliation(s)
- Monireh Hajimoradi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zuhair Mohammad Hassan
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahdieh Bakhshi
- Department of Molecular Medicine, Faculty of Advanced Medical Technologies, Gorgan University of Medical Sciences, Gorgan, Iran
| | - Javad Firouzi
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fazel Sahraneshin Samani
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
48
|
Gilloteaux J, Lau HL, Gourari I, Neal D, Jamison JM, Summers J. Apatone ® induces endometrioid ovarian carcinoma (MDAH 2774) cells to undergo karyolysis and cell death by autoschizis: A potent and safe anticancer treatment. TRANSLATIONAL RESEARCH IN ANATOMY 2015. [DOI: 10.1016/j.tria.2015.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
49
|
Huang C, Wang L, Yang X, Lai L, Chen D, Duan C. Expression of activated signal transducer and activator of transcription-3 as a predictive and prognostic marker in advanced esophageal squamous cell carcinoma. World J Surg Oncol 2015; 13:314. [PMID: 26553224 PMCID: PMC4640156 DOI: 10.1186/s12957-015-0726-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 10/28/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Signal transducer and activator of transcription-3 (STAT3) is an oncogenic transcription factor constitutively active and aberrantly expressed in various types of malignancies, and the expression of p-STAT3 has been recognized as a predictor of poor survival. It remains unclear how variations in p-STAT3 expression influence clinical outcomes in esophageal squamous cell carcinoma (ESCC). METHODS Between 1 January 2008 and 1 November 2013, 153 advanced esophageal squamous cell carcinoma patients (stage IV) from two cancer centers in West China were treated with paclitaxel and cisplatin. We retrospectively analyzed the clinical outcomes of patients with ESCC and examined the correlation between p-STAT3 levels and clinical outcomes in esophageal cancer patients. RESULTS Among the 153 patients, positive p-STAT3 expression was observed in 73 of 153 (47.7 %) cases. The median PFS for patients with positive expression of p-STAT3 and negative expression of p-STAT3 was 5.0 months and 6.9 months, respectively (P < 0.001). The median overall survival was significantly higher in patients with p-STAT3 negative tumors than in those with p-STAT3 positive tumors (9.9 vs 8.9 months, P = 0.026). Kaplan-Meier survival analysis showed that p-STAT3 expression was statistically indicative of a poor prognosis for progression-free survival. CONCLUSIONS These data showed that p-STAT3 expression was significantly associated with poor prognosis in patients with esophageal cancer and could be used as a predictive and prognostic marker in esophageal cancer.
Collapse
Affiliation(s)
- Chuying Huang
- Department of Respiration, Affiliated Hospital of Hubei Institute for Nationalities, Enshi, Hubei Province, China. .,Department of Medical Oncology, Enshi Tujia and Miao Autonomous Prefecture Central Hospital, No. 158, Wu Yang Avenue, Enshi, Hubei Province, China.
| | - Li Wang
- Department of Dermatology, Enshi Tujia and Miao Autonomous Prefecture Central Hospital, No. 158, Wu Yang Avenue, Enshi, Hubei Province, China.
| | - Xibiao Yang
- Department of Radiology, West China Hospital of Sichuan University, No. 37, Guo Xue Xiang, Chengdu, Sichuan, China.
| | - Lin Lai
- Department of Medical Oncology, Enshi Tujia and Miao Autonomous Prefecture Central Hospital, No. 158, Wu Yang Avenue, Enshi, Hubei Province, China.
| | - Dian Chen
- Department of Medical Oncology, Enshi Tujia and Miao Autonomous Prefecture Central Hospital, No. 158, Wu Yang Avenue, Enshi, Hubei Province, China.
| | - Chunyan Duan
- Department of Medical Oncology, Enshi Tujia and Miao Autonomous Prefecture Central Hospital, No. 158, Wu Yang Avenue, Enshi, Hubei Province, China.
| |
Collapse
|
50
|
Abroun S, Saki N, Ahmadvand M, Asghari F, Salari F, Rahim F. STATs: An Old Story, Yet Mesmerizing. CELL JOURNAL 2015; 17:395-411. [PMID: 26464811 PMCID: PMC4601860 DOI: 10.22074/cellj.2015.1] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 08/07/2014] [Indexed: 01/01/2023]
Abstract
Signal transducers and activators of transcription (STATs) are cytoplasmic transcription factors that have a key role in cell fate. STATs, a protein family comprised of
seven members, are proteins which are latent cytoplasmic transcription factors that
convey signals from the cell surface to the nucleus through activation by cytokines
and growth factors. The signaling pathways have diverse biological functions that
include roles in cell differentiation, proliferation, development, apoptosis, and inflammation which place them at the center of a very active area of research. In this review we explain Janus kinase (JAK)/STAT signaling and focus on STAT3, which is
transient from cytoplasm to nucleus after phosphorylation. This procedure controls
fundamental biological processes by regulating nuclear genes controlling cell proliferation, survival, and development. In some hematopoietic disorders and cancers,
overexpression and activation of STAT3 result in high proliferation, suppression of
cell differentiation and inhibition of cell maturation. This article focuses on STAT3
and its role in malignancy, in addition to the role of microRNAs (miRNAs) on STAT3
activation in certain cancers.
Collapse
Affiliation(s)
- Saeid Abroun
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Najmaldin Saki
- Health Research Institute, Research Center of Thalassemia and Hemoglobinopathy, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Ahmadvand
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Farahnaz Asghari
- Department of Medicine II, Division of Gastroenterology, University of Rostock, E.Heydemann-Strasse 6, Rostock, Germany
| | - Fatemeh Salari
- Health Research Institute, Research Center of Thalassemia and Hemoglobinopathy, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fakher Rahim
- Health Research Institute, Hearing Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|