151
|
Fernandes GFDS, Fernandes BC, Valente V, Dos Santos JL. Recent advances in the discovery of small molecules targeting glioblastoma. Eur J Med Chem 2018; 164:8-26. [PMID: 30583248 DOI: 10.1016/j.ejmech.2018.12.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/22/2022]
Abstract
Glioblastoma (GBM) is one of the most common central nervous system cancers. It is characterized as a fast-growing tumor that arises from multiple cell types with neural stem-cell-like properties. Additionally, GBM tumors are highly invasive, which is attributed to the presence of glioblastoma stem cells that makes surgery ineffective in most cases. Currently, temozolomide is the unique chemotherapy option approved by the U.S. Food and Drug Administration for GBM treatment. This review analyzes the emergence and development of new synthetic small molecules discovered as promising anti-glioblastoma agents. A number of compounds were described herein and grouped according to the main chemical class used in the drug discovery process. Importantly, we focused only on synthetic compounds published in the last 10 years, thus excluding natural products. Furthermore, we included in this review only those most biologically active compounds with proven in vitro and/or in vivo efficacy.
Collapse
Affiliation(s)
- Guilherme Felipe Dos Santos Fernandes
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil; São Paulo State University (UNESP), Institute of Chemistry, Araraquara, 14800-060, Brazil
| | - Barbara Colatto Fernandes
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil
| | - Valeria Valente
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil
| | - Jean Leandro Dos Santos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil; São Paulo State University (UNESP), Institute of Chemistry, Araraquara, 14800-060, Brazil.
| |
Collapse
|
152
|
Nabavi SM, Ahmed T, Nawaz M, Devi KP, Balan DJ, Pittalà V, Argüelles-Castilla S, Testai L, Khan H, Sureda A, de Oliveira MR, Vacca RA, Xu S, Yousefi B, Curti V, Daglia M, Sobarzo-Sánchez E, Filosa R, Nabavi SF, Majidinia M, Dehpour AR, Shirooie S. Targeting STATs in neuroinflammation: The road less traveled! Pharmacol Res 2018; 141:73-84. [PMID: 30550953 DOI: 10.1016/j.phrs.2018.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/01/2018] [Accepted: 12/10/2018] [Indexed: 12/16/2022]
Abstract
JAK/STAT transduction pathway is a highly conserved pathway implicated in regulating cellular proliferation, differentiation, survival and apoptosis. Dysregulation of this pathway is involved in the onset of autoimmune, haematological, oncological, metabolic and neurological diseases. Over the last few years, the research of anti-neuroinflammatory agents has gained considerable attention. The ability to diminish the STAT-induced transcription of inflammatory genes is documented for both natural compounds (such as polyphenols) and chemical drugs. Among polyphenols, quercetin and curcumin directly inhibit STAT, while Berberis vulgaris L. and Sophora alopecuroides L extracts act indirectly. Also, the Food and Drug Administration has approved several JAK/STAT inhibitors (direct or indirect) for treating inflammatory diseases, indicating STAT can be considered as a therapeutic target for neuroinflammatory pathologies. Considering the encouraging data obtained so far, clinical trials are warranted to demonstrate the effectiveness and potential use in the clinical practice of STAT inhibitors to treat inflammation-associated neurodegenerative pathologies.
Collapse
Affiliation(s)
- Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Touqeer Ahmed
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad, 44000, Pakistan
| | - Maheen Nawaz
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad, 44000, Pakistan
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University (Science Campus), Karaikudi 630 003, Tamil Nadu, India
| | - Devasahayam Jaya Balan
- Department of Biotechnology, Alagappa University (Science Campus), Karaikudi 630 003, Tamil Nadu, India
| | - Valeria Pittalà
- Department of Drug Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | | | - Lara Testai
- Department of Pharmacy, University of Pisa, Pisa, via Bonanno 6 - 56126, Pisa, Italy
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress and CIBEROBN (Physiopathology of Obesity and Nutrition), University of Balearic Islands, E-07122 Palma de Mallorca, Spain.
| | - Marcos Roberto de Oliveira
- Department of Chemistry/ICET, Federal University of Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, 78060-900, Brazil
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, I-70126, Bari, Italy
| | - Suowen Xu
- University of Rochester, Aab Cardiovascular Research Institute, Rochester, NY, 14623, USA
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Valeria Curti
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Maria Daglia
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Eduardo Sobarzo-Sánchez
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782, Spain; Instituto de Investigación e Innovación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago, Chile
| | - Rosanna Filosa
- Consorzio Sannio Tech, Appia Str, Apollosa, BN 82030, Italy
| | - Seyed Fazel Nabavi
- Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran; Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Shirooie
- Department of Pharmacology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
153
|
Chun J, Song K, Kim YS. Sesquiterpene lactones-enriched fraction of Inula helenium L. induces apoptosis through inhibition of signal transducers and activators of transcription 3 signaling pathway in MDA-MB-231 breast cancer cells. Phytother Res 2018; 32:2501-2509. [PMID: 30251272 DOI: 10.1002/ptr.6189] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/15/2018] [Accepted: 08/15/2018] [Indexed: 11/12/2022]
Abstract
Inula helenium L., commonly known as Elecampane, has been extensively used for many countries in the folk medicine. Its root is a rich source of sesquiterpene lactones, which possess various pharmacological activities. To develop the phytomedicine including sesquiterpene lactones, we prepared hexane fraction from I. helenium (HFIH) and examined the inhibitory effect of HFIH on signal transducers and activators of transcription 3 (STAT3) activation in human breast cancer MDA-MB-231 cells. Additionally, detailed chemical investigation was done to pinpoint the most active sesquiterpene lactones responsible for its anticancer activity. HFIH selectively suppressed STAT3 phosphorylation at tyrosine 705, not affecting its upstream kinases. HFIH downregulated the expression of STAT3 target genes including cyclin D1 , c-myc, and bcl-2 and induced caspase-mediated apoptosis. Moreover, sesquiterpene lactones of HFIH clearly suppressed STAT3 activation. The in vivo results further supported that HFIH inhibits the growth of human breast xenograft tumors. Our results suggest that HFIH possesses potential anticancer activity, which is mainly mediated through STAT3 signaling pathway. These findings provide the potential of HFIH as a promising phytomedicine for the treatment and prevention of triple-negative breast cancer.
Collapse
Affiliation(s)
- Jaemoo Chun
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Kwangho Song
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yeong Shik Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
154
|
Clotaire DZJ, Du X, Wei Y, Yang D, Hua J. miR-19b-3p integrates Jak-Stat signaling pathway through Plzf to regulate self-renewal in dairy goat male germline stem cells. Int J Biochem Cell Biol 2018; 105:104-114. [PMID: 30393202 DOI: 10.1016/j.biocel.2018.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/19/2018] [Accepted: 10/25/2018] [Indexed: 12/27/2022]
Abstract
Jak-Stat pathway is the first pathway identified to stimulate spermatogonial stem cells (SSCs) self-renewal and maintenance activity. Recent studies have showed that stat3 a crucial gene implicated in this pathway can regulate self-renewal in male germline stem cell. In our previous study, we demonstrated that miR-19b-3p induces cell proliferation and reduces heterochromatin through Plzf which also regulates the balance between cell self-renewal and differentiation. Because miRNA can target several genes and to understand more about Plzf, a crucial transcription factor of SSCs, we performed microarray and found that miR-19b-3p integrate Jak-Stat through Plzf to regulate cell self-renewal. Our results demonstrated that miR-19b-3p induces Jak-Stat when Plzf is downregulated; overexpression of Plzf reversed the trend and shown an existence of feedback (-/+) between Plzf and GHR. The cell self-renewal markers CD49f, GFRα1, Oct4 and cKIT analyzed in the both groups miR-19b-3p and Plzf-overexpressing compared to their respective control confirm miR-19b-3p regulates cell pluripotency and self-renewal in goat male germline stem cells through Plzf. Together our finding revels that miR-19b-3p control Jak-Stat signaling through Plzf.
Collapse
Affiliation(s)
- Daguia Zambe John Clotaire
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China; Laboratoire des sciences Agronomiques et Biologiques pour le Développement (LASBAD), Faculty of Science, University of Bangui, P.O Box 908, Central Africa, Central African Republic
| | - Xiaomin Du
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Yudong Wei
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Donghui Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China.
| |
Collapse
|
155
|
Synthesis and Biological Evaluation of Phaeosphaeride A Derivatives as Antitumor Agents. Molecules 2018; 23:molecules23113043. [PMID: 30469343 PMCID: PMC6278656 DOI: 10.3390/molecules23113043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 11/17/2022] Open
Abstract
New derivatives of phaeosphaeride A (PPA) were synthesized and characterized. Anti-tumor activity studies were carried out on the HCT-116, PC3, MCF-7, A549, К562, NCI-Н929, Jurkat, THP-1, RPMI8228 tumor cell lines, and on the HEF cell line. All of the compounds synthesized were found to have better efficacy than PPA towards the tumor cell lines mentioned. Compound 6 was potent against six cancer cell lines, HCT-116, PC-3, K562, NCI-H929, Jurkat, and RPMI8226, showing a 47, 13.5, 16, 4, 1.5, and 7-fold increase in anticancer activity comparative to those of etoposide, respectively. Compound 1 possessed selectivity toward the NCI-H929 cell line (IC50 = 1.35 ± 0.69 μM), while product 7 was selective against three cancer cell lines, HCT-116, MCF-7, and NCI-H929, each having IC50 values of 1.65 μM, 1.80 μM and 2.00 μM, respectively.
Collapse
|
156
|
Song J, Zhang X, Ge Q, Yuan C, Chu L, Liang H, Liao Z, Liu Q, Zhang Z, Zhang B. CRISPR/Cas9-mediated knockout of HBsAg inhibits proliferation and tumorigenicity of HBV-positive hepatocellular carcinoma cells. J Cell Biochem 2018; 119:8419-8431. [PMID: 29904948 PMCID: PMC6221038 DOI: 10.1002/jcb.27050] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 04/23/2018] [Indexed: 12/20/2022]
Abstract
Chronic hepatitis B virus (HBV) infection remains the most common risk factor for hepatocellular carcinoma (HCC). High HBV surface antigen (HBsAg) levels are highly correlated with hepatocarcinogenesis and HBV-associated HCC development. However, the role and detailed mechanisms associated with HBsAg in HCC development remain elusive. In this study, we designed specific single guide RNAs (sgRNAs) targeting the open reading frames, preS1/preS2/S, of the HBV genome and established HBsAg knockout HCC cell lines using the CRISPR/Cas9 system. We showed that knockout of HBsAg in HCC cell lines decreased HBsAg expression and significantly attenuated HCC proliferation in vitro, as well as tumorigenicity in vivo. We also found that overexpression of HBsAg, including the large (LHBs), middle (MHBs), and small (SHBs) surface proteins promoted proliferation and tumor formation in HCC cells. Moreover, we demonstrated that knockout of HBsAg in HCC cells decreased interleukin (IL)-6 production and inhibited signal transducer and activator of transcription 3 (STAT3) signaling, while overexpression of HBsAg induced a substantial accumulation of pY-STAT3. Collectively, these results highlighted the tumorigenic role of HBsAg and implied that the IL-6-STAT3 pathway may be implicated in the HBsAg-mediated malignant potential of HBV-associated HCC.
Collapse
Affiliation(s)
- Jia Song
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province for the Clinical Medicine Research Center of Hepatic SurgeryWuhanChina
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public HealthWuhanChina
| | - Xiaochao Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province for the Clinical Medicine Research Center of Hepatic SurgeryWuhanChina
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public HealthWuhanChina
| | - Qianyun Ge
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province for the Clinical Medicine Research Center of Hepatic SurgeryWuhanChina
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public HealthWuhanChina
| | - Chaoyi Yuan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province for the Clinical Medicine Research Center of Hepatic SurgeryWuhanChina
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public HealthWuhanChina
| | - Liang Chu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province for the Clinical Medicine Research Center of Hepatic SurgeryWuhanChina
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public HealthWuhanChina
| | - Hui‐fang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province for the Clinical Medicine Research Center of Hepatic SurgeryWuhanChina
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public HealthWuhanChina
| | - Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province for the Clinical Medicine Research Center of Hepatic SurgeryWuhanChina
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public HealthWuhanChina
| | - Qiumeng Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province for the Clinical Medicine Research Center of Hepatic SurgeryWuhanChina
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public HealthWuhanChina
| | - Zhanguo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province for the Clinical Medicine Research Center of Hepatic SurgeryWuhanChina
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public HealthWuhanChina
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province for the Clinical Medicine Research Center of Hepatic SurgeryWuhanChina
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public HealthWuhanChina
| |
Collapse
|
157
|
El Sayed I, Helmy MW, El-Abhar HS. Inhibition of SRC/FAK cue: A novel pathway for the synergistic effect of rosuvastatin on the anti-cancer effect of dasatinib in hepatocellular carcinoma. Life Sci 2018; 213:248-257. [PMID: 30292831 DOI: 10.1016/j.lfs.2018.10.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 09/20/2018] [Accepted: 10/03/2018] [Indexed: 01/01/2023]
Abstract
PURPOSE Statins extended their hypocholestremic effect to show a promising anticancer activity. Hepatocellular carcinoma (HCC), the third common cause of cancer-related death, responded positively to statins. Some in-vitro studies reveal the rosuvastatin antitumor effect, but barely in-vivo studies. Hence, we evaluated the antitumor potential of rosuvastatin in a HCC model, the possible signaling cues involved, and whether it augments the dasatinib anticancer effect. METHOD For the in-vitro study, the IC50 and the combination (CI)/dose reduction (DRI) indices were determined for HCC cell line (HepG2) treated with dasatinib and/or rosuvastatin. For the in-vivo study, mice with diethylnitrosamine-induced HCC were treated for 21 days with dasatinib and/or rosuvastatin (10 and 20 mg/kg, respectively). The p-focal adhesion kinase/p-rous sarcoma oncogene cellular homolog (p-FAK/p-Src) cascade and its downstream molecules were assessed. RESULTS The in-vitro study confirmed the synergistic effect of rosuvastatin with dasatinib, which entailed the in-vivo results. The two drugs decreased the p-FAK/p-Src cue along with p-Ras/c-Raf, p-STAT-3, and p-Akt levels to enhance apoptosis by an increase in caspase-3 level and a decline in survivin level. Additionally, they inhibited HGF, VEGF, and the MMP-9. Moreover, the different treatments downregulated the expression of proliferative cell nuclear antigen (PCNA) and Ki-67. The best effect was mediated by the combination regimen that surpassed the effect of either drug alone. CONCLUSION Our results highlighted some of the signals involved in rosuvastatin antitumor effect and nominate it as an adds-on therapy with dasatinib to yield a better effect in HCC through inhibiting the FAK/Src cascade.
Collapse
Affiliation(s)
- Ibrahim El Sayed
- Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Maged W Helmy
- Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, El-Bahira, Egypt.
| | - Hanan S El-Abhar
- Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
158
|
Arora L, Kumar AP, Arfuso F, Chng WJ, Sethi G. The Role of Signal Transducer and Activator of Transcription 3 (STAT3) and Its Targeted Inhibition in Hematological Malignancies. Cancers (Basel) 2018; 10:cancers10090327. [PMID: 30217007 PMCID: PMC6162647 DOI: 10.3390/cancers10090327] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 12/22/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3), a member of the STAT protein family, can be phosphorylated by receptor-associated Janus kinases (JAKs) in response to stimulation by cytokines and growth factors. It forms homo- or heterodimers that can translocate to the cell nucleus where they act as transcription activators. Constitutive activation of STAT3 has been found to be associated with initiation and progression of various cancers. It can exert proliferative as well as anti-apoptotic effects. This review focuses on the role of STAT3 in pathogenesis i.e., proliferation, differentiation, migration, and apoptosis of hematological malignancies viz. leukemia, lymphoma and myeloma, and briefly highlights the potential therapeutic approaches developed against STAT3 activation pathway.
Collapse
Affiliation(s)
- Loukik Arora
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
- Cancer Science Institute of Singapore, Centre for Translational Medicine, 14 Medical Drive, #11-01M, Singapore 117599, Singapore.
- Medical Science Cluster, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia.
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore.
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| | - Wee Joo Chng
- Cancer Science Institute of Singapore, Centre for Translational Medicine, 14 Medical Drive, #11-01M, Singapore 117599, Singapore.
- Department of Hematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119074, Singapore.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6009, Australia.
| |
Collapse
|
159
|
Overexpressed C14orf166 associates with disease progression and poor prognosis in non-small-cell lung cancer. Biosci Rep 2018; 38:BSR20180479. [PMID: 30126850 PMCID: PMC6137245 DOI: 10.1042/bsr20180479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/12/2018] [Accepted: 08/09/2018] [Indexed: 01/05/2023] Open
Abstract
Chromosome 14 ORF 166 (C14orf166), a protein involved in the regulation of RNA transcription and translation, has been reported to possess the potency to promote tumorigenesis; however, the role of C14orf166 in non-small-cell lung cancer (NSCLC) remains unknown. The purpose of the present study was to assess C14orf166 expression and its clinical significance in NSCLC. Immunohistochemical staining, quantitative real-time PCR (qRT-PCR), and Western blotting were used to detect the C14orf166 protein and mRNA expression levels in NSCLC tissues compared with adjacent normal tissues, as well as in NSCLC cells lines compared with normal human bronchial epithelial cells (HBE). Then, the correlations between the C14orf166 expression levels and the clinicopathological features of NSCLC were analyzed. Additionally, the Cox proportional hazard model was used to evaluate the prognostic significance of C14orf166. We found that C14orf166 expression increased in carcinoma tissues compared with their adjacent normal tissues at the protein (P<0.001) and mRNA levels (P<0.001). High expression of C14orf166 was significantly associated with the T stage (P=0.006), lymph node metastasis (P=0.001), advanced TNM stage (P<0.001), and chemotherapy (P<0.001). Moreover, according to the survival analysis, patients with overexpressed C14orf166 were inclined to experience a shorter overall survival and disease-free survival time (P<0.001). Multivariate COX analysis implied that C14orf166 was an independent prognostic biomarker. Taken together, our findings indicate that the overexpression of C14orf166 may contribute to the disease progression of NSCLC, represent a novel prognostic predictor and help high-risk patients make better decisions for subsequent therapy.
Collapse
|
160
|
Gay S, Bugeon J, Bouchareb A, Henry L, Delahaye C, Legeai F, Montfort J, Le Cam A, Siegel A, Bobe J, Thermes V. MiR-202 controls female fecundity by regulating medaka oogenesis. PLoS Genet 2018; 14:e1007593. [PMID: 30199527 PMCID: PMC6147661 DOI: 10.1371/journal.pgen.1007593] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 09/20/2018] [Accepted: 07/26/2018] [Indexed: 12/12/2022] Open
Abstract
Female gamete production relies on coordinated molecular and cellular processes that occur in the ovary throughout oogenesis. In fish, as in other vertebrates, these processes have been extensively studied both in terms of endocrine/paracrine regulation and protein expression and activity. The role of small non-coding RNAs in the regulation of animal reproduction remains however largely unknown and poorly investigated, despite a growing interest for the importance of miRNAs in a wide variety of biological processes. Here, we analyzed the role of miR-202, a miRNA predominantly expressed in male and female gonads in several vertebrate species. We studied its expression in the medaka ovary and generated a mutant line (using CRISPR/Cas9 genome editing) to determine its importance for reproductive success with special interest for egg production. Our results show that miR-202-5p is the most abundant mature form of the miRNA and that it is expressed in granulosa cells and in the unfertilized egg. The knock out (KO) of mir-202 gene resulted in a strong phenotype both in terms of number and quality of eggs produced. Mutant females exhibited either no egg production or produced a dramatically reduced number of eggs that could not be fertilized, ultimately leading to no reproductive success. We quantified the size distribution of the oocytes in the ovary of KO females and performed a large-scale transcriptomic analysis approach to identified dysregulated molecular pathways. Together, cellular and molecular analyses indicate that the lack of miR-202 impairs the early steps of oogenesis/folliculogenesis and decreases the number of large (i.e. vitellogenic) follicles, ultimately leading to dramatically reduced female fecundity. This study sheds new light on the regulatory mechanisms that control the early steps of follicular development, including possible targets of miR-202-5p, and provides the first in vivo functional evidence that a gonad-predominant microRNA may have a major role in female reproduction. The role of small non-coding RNAs in the regulation of animal reproduction remains poorly investigated, despite a growing interest for the importance of miRNAs in a wide variety of biological processes. Here, we analyzed the role of miR-202, a miRNA predominantly expressed in gonads in vertebrate. We studied its expression in the medaka ovary and knocked out the mir-202 gene to study its importance for reproductive success. We showed that the lack of miR-202 results in the sterility of both females and males. In particular, it led to a drastic reduction of both the number and the quality of eggs produced by females. Mutant females exhibited either no egg production or produced a drastically reduced number of eggs that could not be fertilized, ultimately leading to no reproductive success. Quantitative histological and molecular analyses indicated that mir-202 KO impairs oocyte development and is also associated with the dysregulation of many genes that are critical for reproduction. This study sheds new light on the regulatory mechanisms that control oogenesis, including possible targets of miR-202-5p, and provides the first in vivo functional evidence that a gonad-predominant microRNA may have a major role in female reproduction.
Collapse
Affiliation(s)
| | | | | | | | - Clara Delahaye
- LPGP, INRA, Rennes, France
- Univ Rennes, INRIA, CNRS, IRISA, Rennes, France
| | - Fabrice Legeai
- Univ Rennes, INRIA, CNRS, IRISA, Rennes, France
- IGEPP, INRA BP35327, Le Rheu, France
| | | | | | - Anne Siegel
- Univ Rennes, INRIA, CNRS, IRISA, Rennes, France
| | | | | |
Collapse
|
161
|
Seo JH, Choi HW, Oh HN, Lee MH, Kim E, Yoon G, Cho SS, Park SM, Cho YS, Chae JI, Shim JH. Licochalcone D directly targets JAK2 to induced apoptosis in human oral squamous cell carcinoma. J Cell Physiol 2018; 234:1780-1793. [PMID: 30070696 DOI: 10.1002/jcp.27050] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/26/2018] [Indexed: 01/11/2023]
Abstract
Licochalcone (LC) families have been reported to have a wide range of biological function such as antioxidant, antibacterial, antiviral, and anticancer effects. Although various beneficial effects of LCD were revealed, its anticancer effect in human oral squamous cancer has not been identified. To examine the signaling pathway of LCD's anticancer effect, we determined whether LCD has physical interaction with Janus kinase (JAK2)/signal transducer and activator of transcription-3 (STAT3) signaling, which is critical in promoting cancer cell survival and proliferation. Our results demonstrated that LCD inhibited the kinase activity of JAK2, soft agar colony formation, and the proliferation of HN22 and HSC4 cells. LCD also induced mitochondrial apoptotic events such as altered mitochondrial membrane potential and reactive oxygen species production. LCD increased the expression of apoptosis-associated proteins in oral squamous cell carcinoma (OSCC) cells. Finally, the xenograft study showed that LCD significantly inhibited HN22 tumor growth. Immunohistochemical data supported that LCD suppressed p-JAK2 and p-STAT3 expression and induced cleaved-caspase-3 expression. These results indicate that the anticancer effect of LCD is due to the direct targeting of JAK2 kinase. Therefore, LCD can be used for therapeutic application against OSCC.
Collapse
Affiliation(s)
- Ji-Hye Seo
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju, Republic of Korea
| | - Hyun Woo Choi
- Department of Animal Science, Chonbuk National University, Jeonju, Republic of Korea
| | - Ha-Na Oh
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan County, Republic of Korea
| | - Mee-Hyun Lee
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Eunae Kim
- College of Pharmacy, Chosun University, Gwangju, Republic of Korea
| | - Goo Yoon
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan County, Republic of Korea
| | - Seung-Sik Cho
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan County, Republic of Korea
| | - Seon-Min Park
- Pohang Center for Evaluation of Biomaterials, Pohang, Gyeongbuk, Republic of Korea
| | - Young Sik Cho
- Department of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Jung-Il Chae
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju, Republic of Korea
| | - Jung-Hyun Shim
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan County, Republic of Korea.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| |
Collapse
|
162
|
Egawa T, Ohno Y, Yokoyama S, Goto A, Ito R, Hayashi T, Goto K. The effect of advanced glycation end products on cellular signaling molecules in skeletal muscle. THE JOURNAL OF PHYSICAL FITNESS AND SPORTS MEDICINE 2018. [DOI: 10.7600/jpfsm.7.229] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Tatsuro Egawa
- Laboratory of Health and Exercise Sciences, Graduate School of Human and Environmental Studies, Kyoto University
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental Studies, Kyoto University
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University
| | - Yoshitaka Ohno
- Laboratory of Physiology, School of Health Sciences, Toyohashi SOZO University
| | - Shingo Yokoyama
- Laboratory of Physiology, School of Health Sciences, Toyohashi SOZO University
| | - Ayumi Goto
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental Studies, Kyoto University
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University
| | - Rika Ito
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University
| | - Tatsuya Hayashi
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental Studies, Kyoto University
| | - Katsumasa Goto
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University
- Laboratory of Physiology, School of Health Sciences, Toyohashi SOZO University
| |
Collapse
|
163
|
Yang IH, Jung W, Kim LH, Shin JA, Cho NP, Hong SD, Hong KO, Cho SD. Nitidine chloride represses Mcl-1 protein via lysosomal degradation in oral squamous cell carcinoma. J Oral Pathol Med 2018; 47:823-829. [PMID: 29924888 DOI: 10.1111/jop.12755] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/15/2018] [Accepted: 06/18/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND We have shown previously that nitidine chloride (NC) induces apoptosis via inhibition of signal transducer and activator of transcription 3 (STAT3). However, its downstream molecules are not fully understood yet. Here, we report that NC as STAT3 inhibitor downregulates myeloid cell leukemia-1 (Mcl-1) protein in HSC-3 and HSC-4 human oral squamous cell carcinoma (OSCC) cells and a nude mouse tumor xenograft model. METHODS This study investigated the effects of NC on Mcl-1 expression in HSC-3 and HSC-4 cells using Western blotting, RT-PCR, and dual-luciferase assay. Immunohistochemistry was employed to evaluate Mcl-1 expression levels in mouse tumor tissues. Construction of Mcl-1 overexpression vector and transient transfection was done to test the apoptosis of HSC-3 cells. RESULTS Nitidine chloride did not affect either mRNA level or promoter activity of Mcl-1, and the decrease in Mcl-1 protein by NC was caused by lysosome-dependent degradation, but not proteasome-dependent degradation. The overexpression of Mcl-1 protein in OSCC cell lines was sufficient to block the induction of apoptosis. In addition, NC strongly reduced the expression level of Mcl-1 protein compared with other STAT3 inhibitors such as cryptotanshione and S3I-201 in OSCCs. CONCLUSIONS Our findings suggest that NC triggers apoptosis via lysosome-dependent Mcl-1 protein degradation and could be chosen as a promising chemotherapeutic candidate against human OSCCs.
Collapse
Affiliation(s)
- In-Hyoung Yang
- Department of Oral Pathology, School of Dentistry, Institute of Biodegradable Material, Institute of Oral Bioscience, Chonbuk National University, Jeonju, Korea
| | - Won Jung
- Department of Oral medicine, School of Dentistry, Chonbuk National University, Jeonju, Korea
| | - Lee-Han Kim
- Department of Oral Pathology, School of Dentistry, Institute of Biodegradable Material, Institute of Oral Bioscience, Chonbuk National University, Jeonju, Korea
| | - Ji-Ae Shin
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Nam-Pyo Cho
- Department of Oral Pathology, School of Dentistry, Institute of Biodegradable Material, Institute of Oral Bioscience, Chonbuk National University, Jeonju, Korea
| | - Seong Doo Hong
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Kyoung-Ok Hong
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
164
|
Chang Z, Wang Y, Zhou X, Long JE. STAT3 roles in viral infection: antiviral or proviral? Future Virol 2018; 13:557-574. [PMID: 32201498 PMCID: PMC7079998 DOI: 10.2217/fvl-2018-0033] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a transcription factor which can be activated by cytokines, growth factor receptors, and nonreceptor-like tyrosine kinase. An activated STAT3 translocates into the nucleus and combines with DNA to regulate the expression of target genes involved in cell proliferation, differentiation, apoptosis and metastasis. Recent studies have shown that STAT3 plays important roles in viral infection and pathogenesis. STAT3 exhibits a proviral function in several viral infections, including those of HBV, HCV, HSV-1, varicella zoster virus, human CMV and measles virus. However, in some circumstances, STAT3 has an antiviral function in other viral infections, such as enterovirus 71, severe acute respiratory syndrome coronavirus and human metapneumovirus. This review summarizes the roles of STAT3 in viral infection and pathogenesis, and briefly discusses the molecular mechanisms involved in these processes.
Collapse
Affiliation(s)
- Zhangmei Chang
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Kunshan Center For Disease Control & Prevention, 458 Tongfengxi Road, Kunshan, Jiangsu, 215301, PR China.,Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Kunshan Center For Disease Control & Prevention, 458 Tongfengxi Road, Kunshan, Jiangsu, 215301, PR China
| | - Yan Wang
- Department of Medical Microbiology & Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan R., Shanghai 200032, PR China.,Department of Medical Microbiology & Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan R., Shanghai 200032, PR China
| | - Xin Zhou
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | - Jian-Er Long
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Department of Medical Microbiology & Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan R., Shanghai 200032, PR China.,Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Department of Medical Microbiology & Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan R., Shanghai 200032, PR China
| |
Collapse
|
165
|
Rotroff DM, Yee SW, Zhou K, Marvel SW, Shah HS, Jack JR, Havener TM, Hedderson MM, Kubo M, Herman MA, Gao H, Mychaleckyi JC, McLeod HL, Doria A, Giacomini KM, Pearson ER, Wagner MJ, Buse JB, Motsinger-Reif AA. Genetic Variants in CPA6 and PRPF31 Are Associated With Variation in Response to Metformin in Individuals With Type 2 Diabetes. Diabetes 2018; 67:1428-1440. [PMID: 29650774 PMCID: PMC6014560 DOI: 10.2337/db17-1164] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 04/02/2018] [Indexed: 12/24/2022]
Abstract
Metformin is the first-line treatment for type 2 diabetes (T2D). Although widely prescribed, the glucose-lowering mechanism for metformin is incompletely understood. Here, we used a genome-wide association approach in a diverse group of individuals with T2D from the Action to Control Cardiovascular Risk in Diabetes (ACCORD) clinical trial to identify common and rare variants associated with HbA1c response to metformin treatment and followed up these findings in four replication cohorts. Common variants in PRPF31 and CPA6 were associated with worse and better metformin response, respectively (P < 5 × 10-6), and meta-analysis in independent cohorts displayed similar associations with metformin response (P = 1.2 × 10-8 and P = 0.005, respectively). Previous studies have shown that PRPF31(+/-) knockout mice have increased total body fat (P = 1.78 × 10-6) and increased fasted circulating glucose (P = 5.73 × 10-6). Furthermore, rare variants in STAT3 associated with worse metformin response (q <0.1). STAT3 is a ubiquitously expressed pleiotropic transcriptional activator that participates in the regulation of metabolism and feeding behavior. Here, we provide novel evidence for associations of common and rare variants in PRPF31, CPA6, and STAT3 with metformin response that may provide insight into mechanisms important for metformin efficacy in T2D.
Collapse
Affiliation(s)
- Daniel M Rotroff
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC
- Department of Statistics, North Carolina State University, Raleigh, NC
| | - Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA
| | - Kaixin Zhou
- School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland
| | - Skylar W Marvel
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC
| | - Hetal S Shah
- Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - John R Jack
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC
| | - Tammy M Havener
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | - Michiaki Kubo
- RIKEN Center for Integrative Medical Science, Yokohama, Japan
| | | | - He Gao
- Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - Josyf C Mychaleckyi
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA
| | | | | | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA
| | - Ewan R Pearson
- School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland
| | - Michael J Wagner
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - John B Buse
- Division of Endocrinology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Alison A Motsinger-Reif
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC
- Department of Statistics, North Carolina State University, Raleigh, NC
| | | | | |
Collapse
|
166
|
Yuza K, Nagahashi M, Shimada Y, Nakano M, Tajima Y, Kameyama H, Nakajima M, Takabe K, Wakai T. Upregulation of phosphorylated sphingosine kinase 1 expression in colitis-associated cancer. J Surg Res 2018; 231:323-330. [PMID: 30278948 DOI: 10.1016/j.jss.2018.05.085] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/08/2018] [Accepted: 05/31/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND Colitis-associated cancer (CAC) is the most serious complication of inflammatory bowel disease. Sphingosine-1-phosphate (S1P) is a bioactive lipid mediator that is generated by sphingosine kinase 1 (SphK1) and is known to play an important role in inflammation and cancer progression. Moreover, SphK1 and S1P act as upstream mediators of proinflammatory cytokine interleukin 6 (IL-6) and signal transducer and activator of transcription-3 (STAT3). We hypothesized that the expression levels of phosphorylated SphK1 (pSphK1), phosphorylated STAT3 (pSTAT3), and IL-6 are universally higher in CAC patients than in sporadic colorectal cancer (CRC) patients because all of these factors are associated with inflammation. In this study, we determined the expression levels of pSphK1 in patients with sporadic CRC and CAC and clarified the importance of S1P in CAC patients. MATERIALS AND METHODS We randomly selected 10 sporadic CRC patients and 10 CAC patients who underwent curative resection, and we examined their surgical specimens by immunohistochemistry. We determined the expression levels of pSphK1, pSTAT3, and IL-6 in these samples. RESULTS We found pSphK1 expression to be more prevalent in CAC patients (P = 0.019) and to have a higher immunohistochemistry score (P = 0.005) than in sporadic CRC patients. However, the expression of pSTAT3 and IL-6 did not differ between the patient groups. CONCLUSIONS To our knowledge, this is the first report comparing pSphK1 expression levels in CAC with those in sporadic CRC. The high levels of pSphK1 expression in CAC suggest an important role of S1P in the disease process of CAC.
Collapse
Affiliation(s)
- Kizuki Yuza
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Japan
| | - Masayuki Nagahashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Japan.
| | - Yoshifumi Shimada
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Japan
| | - Mae Nakano
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Japan
| | - Yosuke Tajima
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Japan
| | - Hitoshi Kameyama
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Japan
| | - Masato Nakajima
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Japan
| | - Kazuaki Takabe
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Japan; Breast Surgery, Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, New York; Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, New York; Department of Breast Surgery and Oncology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan; Department of Surgery, Yokohama City University, Kanazawa-ku, Yokohama, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Japan
| |
Collapse
|
167
|
Mohammadian J, Molavi O, Pirouzpanah MB, Rahimi AAR, Samadi N. Stattic enhances the anti-proliferative effect of docetaxel via the Bax/Bcl-2/cyclin B axis in human cancer cells. Process Biochem 2018. [DOI: 10.1016/j.procbio.2018.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
168
|
Lee DS, O'Keefe RA, Ha PK, Grandis JR, Johnson DE. Biochemical Properties of a Decoy Oligodeoxynucleotide Inhibitor of STAT3 Transcription Factor. Int J Mol Sci 2018; 19:ijms19061608. [PMID: 29848966 PMCID: PMC6032396 DOI: 10.3390/ijms19061608] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 05/29/2018] [Indexed: 12/13/2022] Open
Abstract
Cyclic STAT3 decoy (CS3D) is a second-generation, double-stranded oligodeoxynucleotide (ODN) that mimics a genomic response element for signal transducer and activator of transcription 3 (STAT3), an oncogenic transcription factor. CS3D competitively inhibits STAT3 binding to target gene promoters, resulting in decreased expression of proteins that promote cellular proliferation and survival. Previous studies have demonstrated antitumor activity of CS3D in preclinical models of solid tumors. However, prior to entering human clinical trials, the efficiency of generating the CS3D molecule and its stability in biological fluids should be determined. CS3D is synthesized as a single-stranded ODN and must have its free ends ligated to generate the final cyclic form. In this study, we report a ligation efficiency of nearly 95 percent. The ligated CS3D demonstrated a half-life of 7.9 h in human serum, indicating adequate stability for intravenous delivery. These results provide requisite biochemical characterization of CS3D that will inform upcoming clinical trials.
Collapse
Affiliation(s)
- David S Lee
- School of Medicine, University of California at San Francisco, San Francisco, CA 94115, USA.
| | - Rachel A O'Keefe
- Department of Otolaryngology-Head and Neck Surgery, University of California at San Francisco, San Francisco, CA 94115, USA.
| | - Patrick K Ha
- Department of Otolaryngology-Head and Neck Surgery, University of California at San Francisco, San Francisco, CA 94115, USA.
| | - Jennifer R Grandis
- Department of Otolaryngology-Head and Neck Surgery, University of California at San Francisco, San Francisco, CA 94115, USA.
| | - Daniel E Johnson
- Department of Otolaryngology-Head and Neck Surgery, University of California at San Francisco, San Francisco, CA 94115, USA.
| |
Collapse
|
169
|
Zeng Q, Fu J, Korrer M, Gorbounov M, Murray PJ, Pardoll D, Masica DL, Kim YJ. Caspase-1 from Human Myeloid-Derived Suppressor Cells Can Promote T Cell-Independent Tumor Proliferation. Cancer Immunol Res 2018; 6:566-577. [PMID: 29653983 DOI: 10.1158/2326-6066.cir-17-0543] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/02/2018] [Accepted: 03/26/2018] [Indexed: 11/16/2022]
Abstract
Immunosuppressive myeloid-derived suppressive cells (MDSCs) are characterized by their phenotypic and functional heterogeneity. To better define their T cell-independent functions within the tumor, sorted monocytic CD14+CD11b+HLA-DRlow/- MDSCs (mMDSC) from squamous cell carcinoma patients showed upregulated caspase-1 activity, which was associated with increased IL1β and IL18 expression. In vitro studies demonstrated that mMDSCs promoted caspase-1-dependent proliferation of multiple squamous carcinoma cell lines in both human and murine systems. In vivo, growth rates of B16, MOC1, and Panc02 were significantly blunted in chimeric mice adoptively transferred with caspase-1 null bone marrow cells under T cell-depleted conditions. Adoptive transfer of wild-type Gr-1+CD11b+ MDSCs from tumor-bearing mice reversed this antitumor response, whereas caspase-1 inhibiting thalidomide-treated MDSCs phenocopied the antitumor response found in caspase-1 null mice. We further hypothesized that MDSC caspase-1 activity could promote tumor-intrinsic MyD88-dependent carcinogenesis. In mice with wild-type caspase-1, MyD88-silenced tumors displayed reduced growth rate, but in chimeric mice with caspase-1 null bone marrow cells, MyD88-silenced tumors did not display differential tumor growth rate. When we queried the TCGA database, we found that caspase-1 expression is correlated with overall survival in squamous cell carcinoma patients. Taken together, our findings demonstrated that caspase-1 in MDSCs is a direct T cell-independent mediator of tumor proliferation. Cancer Immunol Res; 6(5); 566-77. ©2018 AACR.
Collapse
Affiliation(s)
- Qi Zeng
- Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Juan Fu
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Baltimore, Maryland
| | - Michael Korrer
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Peter J Murray
- Johns Hopkins Hospital, Baltimore, Maryland; Max Planck Institute of Biochemistry, Munich, Germany
| | - Drew Pardoll
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Baltimore, Maryland
| | - David L Masica
- Department of Biomedical Engineering and Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Young J Kim
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, Tennessee. .,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
170
|
Qian P, Zhang YW, Zhou ZH, Liu JQ, Yue SY, Guo XL, Sun LQ, Lv XT, Chen JQ. Artesunate enhances γδ T-cell-mediated antitumor activity through augmenting γδ T-cell function and reversing immune escape of HepG2 cells. Immunopharmacol Immunotoxicol 2018; 40:107-116. [PMID: 29405080 DOI: 10.1080/08923973.2017.1386212] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Peng Qian
- Department of Gastroenterology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yong-Wen Zhang
- Department of Gastroenterology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhong-Hai Zhou
- Department of Central Laboratory, 97th Hospital of PLA, Xuzhou, Jiangsu, China
| | - Jun-Quan Liu
- Department of Central Laboratory, 97th Hospital of PLA, Xuzhou, Jiangsu, China
| | - Su-Yang Yue
- Department of Gastroenterology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiang-Li Guo
- Department of Gastroenterology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lei-Qing Sun
- Department of Central Laboratory, 97th Hospital of PLA, Xuzhou, Jiangsu, China
| | - Xiao-Ting Lv
- Department of Central Laboratory, 97th Hospital of PLA, Xuzhou, Jiangsu, China
| | - Jian-Qun Chen
- Department of Gastroenterology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
171
|
Xu BQ, Fu ZG, Meng Y, Wu XQ, Wu B, Xu L, Jiang JL, Li L, Chen ZN. Gemcitabine enhances cell invasion via activating HAb18G/CD147-EGFR-pSTAT3 signaling. Oncotarget 2018; 7:62177-62193. [PMID: 27556697 PMCID: PMC5308719 DOI: 10.18632/oncotarget.11405] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 08/08/2016] [Indexed: 12/20/2022] Open
Abstract
Pancreatic cancer, one of the most lethal cancers, has very poor 5-year survival partly due to gemcitabine resistance. Recently, it was reported that chemotherapeutic agents may act as stressors to induce adaptive responses and to promote chemoresistance in cancer cells. During long-term drug treatment, the minority of cancer cells survive and acquire an epithelial-mesenchymal transition phenotype with increased chemo-resistance and metastasis. However, the short-term response of most cancer cells remains unclear. This study aimed to investigate the short-term response of pancreatic cancer cells to gemcitabine stress and to explore the corresponding mechanism. Our results showed that gemcitabine treatment for 24 hours enhanced pancreatic cancer cell invasion. In gemcitabine-treated cells, HAb18G/CD147 was up-regulated; and HAb18G/CD147 down-regulation or inhibition attenuated gemcitabine-enhanced invasion. Mechanistically, HAb18G/CD147 promoted gemcitabine-enhanced invasion by activating the EGFR (epidermal growth factor receptor)-STAT3 (signal transducer and activator of transcription 3) signaling pathway. Inhibition of EGFR-STAT3 signaling counteracted gemcitabine-enhanced invasion, and which relied on HAb18G/CD147 levels. In pancreatic cancer tissues, EGFR was highly expressed and positively correlated with HAb18G/CD147. These data indicate that pancreatic cancer cells enhance cell invasion via activating HAb18G/CD147-EGFR-pSTAT3 signaling. Our findings suggest that inhibiting HAb18G/CD147 is a potential strategy for overcoming drug stress-associated resistance in pancreatic cancer.
Collapse
Affiliation(s)
- Bao-Qing Xu
- Department of Cell Biology and Cell Engineering Research Center, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Zhi-Guang Fu
- Department of Cell Biology and Cell Engineering Research Center, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Yao Meng
- Department of Cell Biology and Cell Engineering Research Center, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Xiao-Qing Wu
- Departments of Molecular Biosciences and Radiation Oncology, University of Kansas, Lawrence, Kansas, USA
| | - Bo Wu
- Department of Cell Biology and Cell Engineering Research Center, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Liang Xu
- Departments of Molecular Biosciences and Radiation Oncology, University of Kansas, Lawrence, Kansas, USA
| | - Jian-Li Jiang
- Department of Cell Biology and Cell Engineering Research Center, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Ling Li
- Department of Cell Biology and Cell Engineering Research Center, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Zhi-Nan Chen
- Department of Cell Biology and Cell Engineering Research Center, State Key Laboratory of Cancer Biology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
172
|
Zhu ZY, Jia CZ, Luo JM, Wang L. Polyriboinosinic-polyribocytidylic acid facilitates interleukin-6, and interleukin-8 secretion in human dermal fibroblasts via the JAK/STAT3 and p38 MAPK signal transduction pathways. Cytokine 2018; 102:1-6. [PMID: 29245047 DOI: 10.1016/j.cyto.2017.12.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/07/2017] [Accepted: 12/09/2017] [Indexed: 02/05/2023]
Abstract
Polyriboinosinic-polyribocytidylic acid (polyI:C) is a viral dsRNA analoguethat promotes wounds healing, accelerates re-epithelialization, granulation and neovascularization, and induces pro-inflammatory cytokine release. Little is known about polyI:C mediated induction of inflammatory mediators in human dermal fibroblast (HDFs), which form the primary scaffold for epithelial cells covering the wound. Here, we found that polyI:C enhances IL-6 and IL-8 mRNA expression and induces of IL-6 and IL-8 production in a concentration-dependent and time-dependent manner in HDFs. PolyI:C treatment rapidly increased phosphorylation level of both STAT3 and p38 mitogen-activated protein kinase (MAPK). Moreover, pretreatment with AG490, a Janus kinase (JAK) inhibitor, inhibited polyI:C-induced STAT3 phosphorylation and subsequent IL-6 and IL-8 release. Conversely, pretreatment with SB203580, a selective inhibitor of p38 MAPK, blocked p38 MAPK phosphorylation and IL-6 and IL-8 expression. In conclusion, polyI:C induces IL-6 and IL-8 production in HDFs via the JAK/STAT3 and p38 MAPK signaling pathways.
Collapse
Affiliation(s)
- Zhang Ying Zhu
- Department of Pathophysiology, Shantou University Medical College, 5150412, People's Republic of China
| | - Cong Zhuo Jia
- Department of Dermatology, First Affiliated Hospital, Shantou University Medical College, 515041, People's Republic of China
| | - Jian Min Luo
- Department of Pathophysiology, Shantou University Medical College, 5150412, People's Republic of China.
| | - Li Wang
- Shenzhen University General Hospital, 518055, People's Republic of China; Department of Dermatology, First Affiliated Hospital, Shantou University Medical College, 515041, People's Republic of China; Huizhou Municipal Hospital, People's Republic of China.
| |
Collapse
|
173
|
Bharadwaj U, Eckols TK, Xu X, Kasembeli MM, Chen Y, Adachi M, Song Y, Mo Q, Lai SY, Tweardy DJ. Small-molecule inhibition of STAT3 in radioresistant head and neck squamous cell carcinoma. Oncotarget 2018; 7:26307-30. [PMID: 27027445 PMCID: PMC5041982 DOI: 10.18632/oncotarget.8368] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/14/2016] [Indexed: 12/17/2022] Open
Abstract
While STAT3 has been validated as a target for treatment of many cancers, including head and neck squamous cell carcinoma (HNSCC), a STAT3 inhibitor is yet to enter the clinic. We used the scaffold of C188, a small-molecule STAT3 inhibitor previously identified by us, in a hit-to-lead program to identify C188-9. C188-9 binds to STAT3 with high affinity and represents a substantial improvement over C188 in its ability to inhibit STAT3 binding to its pY-peptide ligand, to inhibit cytokine-stimulated pSTAT3, to reduce constitutive pSTAT3 activity in multiple HNSCC cell lines, and to inhibit anchorage dependent and independent growth of these cells. In addition, treatment of nude mice bearing xenografts of UM-SCC-17B, a radioresistant HNSCC line, with C188-9, but not C188, prevented tumor xenograft growth. C188-9 treatment modulated many STAT3-regulated genes involved in oncogenesis and radioresistance, as well as radioresistance genes regulated by STAT1, due to its potent activity against STAT1, in addition to STAT3. C188-9 was well tolerated in mice, showed good oral bioavailability, and was concentrated in tumors. Thus, C188-9, either alone or in combination with radiotherapy, has potential for use in treating HNSCC tumors that demonstrate increased STAT3 and/or STAT1 activation.
Collapse
Affiliation(s)
- Uddalak Bharadwaj
- Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - T Kris Eckols
- Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xuejun Xu
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, China
| | - Moses M Kasembeli
- Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yunyun Chen
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Makoto Adachi
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yongcheng Song
- Department of Pharmacology, Baylor College of Medicine, Houston, Texas, USA
| | - Qianxing Mo
- Department of Medicine, Division of Biostatistics, Dan L. Duncan Cancer Center, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Stephen Y Lai
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David J Tweardy
- Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Molecular & Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
174
|
The EGFR-ADAM17 Axis in Chronic Obstructive Pulmonary Disease and Cystic Fibrosis Lung Pathology. Mediators Inflamm 2018. [PMID: 29540993 PMCID: PMC5818912 DOI: 10.1155/2018/1067134] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and cystic fibrosis (CF) share molecular mechanisms that cause the pathological symptoms they have in common. Here, we review evidence suggesting that hyperactivity of the EGFR/ADAM17 axis plays a role in the development of chronic lung disease in both CF and COPD. The ubiquitous transmembrane protease A disintegrin and metalloprotease 17 (ADAM17) forms a functional unit with the EGF receptor (EGFR), in a feedback loop interaction labeled the ADAM17/EGFR axis. In airway epithelial cells, ADAM17 sheds multiple soluble signaling proteins by proteolysis, including EGFR ligands such as amphiregulin (AREG), and proinflammatory mediators such as the interleukin 6 coreceptor (IL-6R). This activity can be enhanced by injury, toxins, and receptor-mediated external triggers. In addition to intracellular kinases, the extracellular glutathione-dependent redox potential controls ADAM17 shedding. Thus, the epithelial ADAM17/EGFR axis serves as a receptor of incoming luminal stress signals, relaying these to neighboring and underlying cells, which plays an important role in the resolution of lung injury and inflammation. We review evidence that congenital CFTR deficiency in CF and reduced CFTR activity in chronic COPD may cause enhanced ADAM17/EGFR signaling through a defect in glutathione secretion. In future studies, these complex interactions and the options for pharmaceutical interventions will be further investigated.
Collapse
|
175
|
Li T, Liu X, Shen Q, Yang W, Huo Z, Liu Q, Jiao H, Chen J. Salinomycin exerts anti-angiogenic and anti-tumorigenic activities by inhibiting vascular endothelial growth factor receptor 2-mediated angiogenesis. Oncotarget 2018; 7:26580-92. [PMID: 27058891 PMCID: PMC5042000 DOI: 10.18632/oncotarget.8555] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 03/12/2016] [Indexed: 12/12/2022] Open
Abstract
Anti-angiogenesis targeting VEGFR2 has been an attractive strategy for cancer therapy for its role in promoting cancer growth and metastasis. However, the currently available drugs have unexpected side effects. Therefore, development of novel VEGFR2 inhibitors with less toxicity would be of great value. In this study, we describe a novel and safely VEGFR2 inhibitor, Salinomycin (Sal), which was screened from the drug libraries of Food and Drug Administration (FDA) and prohibited the binding of the ATP at its binding pocket of VEGFR2 using molecular docking model. Sal could interfere a series of VEGF-induced angiogenesis processes including proliferation, migration, and tube formation in HUVECS in vitro. Matrigel plug model demonstrated Sal strongly inhibited angiogenesis in vivo. We found that Sal significantly decreased VEGF-induced phosphorylation of VEGFR2 and its downstream STAT3 in dose- and time-dependent manner in HUVECs. Besides, Sal could directly reduce the cell viability and induce apoptosis in SGC-7901 cancer cells in vitro. Sal inhibited constitutive STAT3 activation by blocking its DNA binding and reduced various gene products including Bcl-2, Bcl-xL and VEGF both at mRNA and protein levels. Intra-peritoneal injection of Sal at doses of 3 and 5 mg/kg/day markedly suppressed human gastric cancer xenografts angiogenesis and growth without causing obvious toxicities. Taken together, Sal inhibits tumor angiogenesis and growth of gastric cancer; our results reveal unique characteristics of Sal as a promising anticancer drug candidate.
Collapse
Affiliation(s)
- Tao Li
- Department of Oncology, General Hospital of the Ningxia Medical University, Yinchuan 750004, China
| | - Xiaoxia Liu
- Department of Medical Genetic and Cell Biology, Ningxia Medical University, Yinchuan 750004, China.,Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan 750004, China
| | - Qin Shen
- Department of Medical Genetic and Cell Biology, Ningxia Medical University, Yinchuan 750004, China.,Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan 750004, China
| | - Wenjun Yang
- Department of Medical Genetic and Cell Biology, Ningxia Medical University, Yinchuan 750004, China.,Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan 750004, China
| | - Zhenghao Huo
- Department of Medical Genetic and Cell Biology, Ningxia Medical University, Yinchuan 750004, China.,Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan 750004, China
| | - Qilun Liu
- Department of Oncology, General Hospital of the Ningxia Medical University, Yinchuan 750004, China
| | - Haiyan Jiao
- Department of Medical Genetic and Cell Biology, Ningxia Medical University, Yinchuan 750004, China.,Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan 750004, China
| | - Jing Chen
- Department of Medical Genetic and Cell Biology, Ningxia Medical University, Yinchuan 750004, China.,Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan 750004, China
| |
Collapse
|
176
|
Popławski P, Wiśniewski JR, Rijntjes E, Richards K, Rybicka B, Köhrle J, Piekiełko-Witkowska A. Restoration of type 1 iodothyronine deiodinase expression in renal cancer cells downregulates oncoproteins and affects key metabolic pathways as well as anti-oxidative system. PLoS One 2017; 12:e0190179. [PMID: 29272308 PMCID: PMC5741248 DOI: 10.1371/journal.pone.0190179] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 12/08/2017] [Indexed: 12/20/2022] Open
Abstract
Type 1 iodothyronine deiodinase (DIO1) contributes to deiodination of 3,5,3’,5’-tetraiodo-L-thyronine (thyroxine, T4) yielding of 3,5,3’-triiodothyronine (T3), a powerful regulator of cell differentiation, proliferation, and metabolism. Our previous work showed that loss of DIO1 enhances proliferation and migration of renal cancer cells. However, the global effects of DIO1 expression in various tissues affected by cancer remain unknown. Here, the effects of stable DIO1 re-expression were analyzed on the proteome of renal cancer cells, followed by quantitative real-time PCR validation in two renal cancer-derived cell lines. DIO1-induced changes in intracellular concentrations of thyroid hormones were quantified by L-MS/MS and correlations between expression of DIO1 and potential target genes were determined in tissue samples from renal cancer patients. Stable re-expression of DIO1, resulted in 26 downregulated proteins while 59 proteins were overexpressed in renal cancer cells. The ‘downregulated’ group consisted mainly of oncoproteins (e.g. STAT3, ANPEP, TGFBI, TGM2) that promote proliferation, migration and invasion. Furthermore, DIO1 re-expression enhanced concentrations of two subunits of thyroid hormone transporter (SLC7A5, SLC3A2), enzymes of key pathways of cellular energy metabolism (e.g. TKT, NAMPT, IDH2), sex steroid metabolism and anti-oxidative response (AKR1C2, AKR1B10). DIO1 expression resulted in elevated intracellular concentration of T4. Expression of DIO1-affected genes strongly correlated with DIO1 transcript levels in tissue samples from renal cancer patients as well as with their poor survival. This first study addressing effects of deiodinase re-expression on proteome of cancer cells demonstrates that induced DIO1 re-expression in renal cancer robustly downregulates oncoproteins, affects key metabolic pathways, and triggers proteins involved in anti-oxidative protection. This data supports the notion that suppressed DIO1 expression and changes in local availability of thyroid hormones might favor a shift from a differentiated to a more proliferation-prone state of cancer tissues and cell lines.
Collapse
Affiliation(s)
- Piotr Popławski
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Jacek R. Wiśniewski
- Biochemical Proteomics Group, Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Eddy Rijntjes
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Keith Richards
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Beata Rybicka
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
177
|
Luo J, Yan R, He X, He J. Constitutive activation of STAT3 and cyclin D1 overexpression contribute to proliferation, migration and invasion in gastric cancer cells. Am J Transl Res 2017; 9:5671-5677. [PMID: 29312519 PMCID: PMC5752917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 11/24/2017] [Indexed: 06/07/2023]
Abstract
Signal transducer and activator of transcription-3 (STAT3) is closely associated with tumorigenesis and is activated in tumor cells of a variety of cancers. The raised expression of cyclin D1 (CCND1) by activated STAT3 has been verified in some cancers. However, the relationship between STAT3 and CCND1 has yet to be studied in gastric cancer (GC) cells. In the present study, we found that STAT3 was constitutively activated in several GC cells together with overexpressed CCND1. In addition, IL-6 treatment enhanced the expression level of p-STAT3 and CCND1, accelerating the cell cycle progress and transferring from G1 to S phase. The increased proliferation, migration and invasion were also demonstrated by the treatment of IL-6 in HGC-27 and BGC-823 cells. While AG490 treatment, a Janus Kinase (JAK) inhibitor, showed the opposite effect. Therefore, our research demonstrated the positive correlation between p-STAT3 and CCND1 in GC cells. The constitutive activation of STAT3 and CCND1 overexpression accounted for the proliferation, migration and invasion in GC cells.
Collapse
Affiliation(s)
- Jianfei Luo
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan UniversityWuhan 430060, Hubei Province, China
| | - Ruicheng Yan
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan UniversityWuhan 430060, Hubei Province, China
| | - Xiaobo He
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan UniversityWuhan 430060, Hubei Province, China
| | - Jie He
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan UniversityWuhan 430060, Hubei Province, China
| |
Collapse
|
178
|
Hu F, Zhao Y, Yu Y, Fang JM, Cui R, Liu ZQ, Guo XL, Xu Q. Docetaxel-mediated autophagy promotes chemoresistance in castration-resistant prostate cancer cells by inhibiting STAT3. Cancer Lett 2017; 416:24-30. [PMID: 29246644 DOI: 10.1016/j.canlet.2017.12.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 12/08/2017] [Accepted: 12/08/2017] [Indexed: 12/20/2022]
Abstract
Signal transducer and activator of transcription (STAT)3 expression is correlated with neoplasm growth, metastasis, and prognosis; it has also been implicated in the regulation of autophagy, which may in turn contribute to tumor chemoresistance. However, it is unknown whether STAT3 is involved in cancer cell survival in response to chemotherapy. In this study, we show that autophagy is triggered during chemotherapy and that inhibiting autophagy increased chemosensitivity of castration-resistant prostate cancer (CRPC) cells. Meanwhile, docetaxel induced autophagy was inhibited by STAT3 activation, which increased mitochondrial damage and decreased CRPC cell viability. These results suggest that STAT3 contributes to CRPC cell survival and chemoresistance by modulating autophagy.
Collapse
Affiliation(s)
- Fei Hu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China
| | - Yu Zhao
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China
| | - Yi Yu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China
| | - Jue-Min Fang
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China
| | - Ran Cui
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China
| | - Zhu-Qing Liu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China
| | - Xian-Ling Guo
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China.
| | - Qing Xu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China.
| |
Collapse
|
179
|
The role of STAT3 in leading the crosstalk between human cancers and the immune system. Cancer Lett 2017; 415:117-128. [PMID: 29222039 DOI: 10.1016/j.canlet.2017.12.003] [Citation(s) in RCA: 262] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/01/2017] [Accepted: 12/01/2017] [Indexed: 12/12/2022]
Abstract
The development and progression of human cancers are continuously and dynamically regulated by intrinsic and extrinsic factors. As a converging point of multiple oncogenic pathways, signal transducer and activator of transcription 3 (STAT3) is constitutively activated both in tumor cells and tumor-infiltrated immune cells. Activated STAT3 persistently triggers tumor progression through direct regulation of oncogenic gene expression. Apart from its oncogenic role in regulating gene expression in tumor cells, STAT3 also paves the way for human cancer growth through immunosuppression. Activated STAT3 in immune cells results in inhibition of immune mediators and promotion of immunosuppressive factors. Therefore, STAT3 modulates the interaction between tumor cells and host immunity. Accumulating evidence suggests that targeting STAT3 may enhance anti-cancer immune responses and rescue the suppressed immunologic microenvironment in tumors. Taken together, STAT3 has emerged as a promising target in cancer immunotherapy.
Collapse
|
180
|
Ko JH, Sethi G, Um JY, Shanmugam MK, Arfuso F, Kumar AP, Bishayee A, Ahn KS. The Role of Resveratrol in Cancer Therapy. Int J Mol Sci 2017; 18:ijms18122589. [PMID: 29194365 PMCID: PMC5751192 DOI: 10.3390/ijms18122589] [Citation(s) in RCA: 485] [Impact Index Per Article: 60.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 12/26/2022] Open
Abstract
Natural product compounds have recently attracted significant attention from the scientific community for their potent effects against inflammation-driven diseases, including cancer. A significant amount of research, including preclinical, clinical, and epidemiological studies, has indicated that dietary consumption of polyphenols, found at high levels in cereals, pulses, vegetables, and fruits, may prevent the evolution of an array of diseases, including cancer. Cancer development is a carefully orchestrated progression where normal cells acquires mutations in their genetic makeup, which cause the cells to continuously grow, colonize, and metastasize to other organs such as the liver, lungs, colon, and brain. Compounds that modulate these oncogenic processes can be considered as potential anti-cancer agents that may ultimately make it to clinical application. Resveratrol, a natural stilbene and a non-flavonoid polyphenol, is a phytoestrogen that possesses anti-oxidant, anti-inflammatory, cardioprotective, and anti-cancer properties. It has been reported that resveratrol can reverse multidrug resistance in cancer cells, and, when used in combination with clinically used drugs, it can sensitize cancer cells to standard chemotherapeutic agents. Several novel analogs of resveratrol have been developed with improved anti-cancer activity, bioavailability, and pharmacokinetic profile. The current focus of this review is resveratrol’s in vivo and in vitro effects in a variety of cancers, and intracellular molecular targets modulated by this polyphenol. This is also accompanied by a comprehensive update of the various clinical trials that have demonstrated it to be a promising therapeutic and chemopreventive agent.
Collapse
Affiliation(s)
- Jeong-Hyeon Ko
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Gautam Sethi
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Jae-Young Um
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth WA 6009, Australia.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA.
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| |
Collapse
|
181
|
Qiu HY, Fu JY, Yang MK, Han HW, Wang PF, Zhang YH, Lin HY, Tang CY, Qi JL, Yang RW, Wang XM, Zhu HL, Yang YH. Identification of new shikonin derivatives as STAT3 inhibitors. Biochem Pharmacol 2017; 146:74-86. [DOI: 10.1016/j.bcp.2017.10.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/19/2017] [Indexed: 01/10/2023]
|
182
|
Wu X, Tang W, Marquez RT, Li K, Highfill CA, He F, Lian J, Lin J, Fuchs JR, Ji M, Li L, Xu L. Overcoming chemo/radio-resistance of pancreatic cancer by inhibiting STAT3 signaling. Oncotarget 2017; 7:11708-23. [PMID: 26887043 PMCID: PMC4905505 DOI: 10.18632/oncotarget.7336] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/23/2016] [Indexed: 12/17/2022] Open
Abstract
Chemo/radio-therapy resistance to the deadly pancreatic cancer is mainly due to the failure to kill pancreatic cancer stem cells (CSCs). Signal transducer and activator of transcription 3 (STAT3) is activated in pancreatic CSCs and, therefore, may be a valid target for overcoming therapeutic resistance. Here we investigated the potential of STAT3 inhibition in sensitizing pancreatic cancer to chemo/radio-therapy. We found that the levels of nuclear pSTAT3 in pancreatic cancer correlated with advanced tumor grade and poor patient outcome. Liposomal delivery of a STAT3 inhibitor FLLL32 (Lip-FLLL32) inhibited STAT3 phosphorylation and STAT3 target genes in pancreatic cancer cells and tumors. Consequently, Lip-FLLL32 suppressed pancreatic cancer cell growth, and exhibited synergetic effects with gemcitabine and radiation treatment in vitro and in vivo. Furthermore, Lip-FLLL32 reduced ALDH1-positive CSC population and modulated several potential stem cell markers. These results demonstrate that Lip-FLLL32 suppresses pancreatic tumor growth and sensitizes pancreatic cancer cells to radiotherapy through inhibition of CSCs in a STAT3-dependent manner. By targeting pancreatic CSCs, Lip-FLLL32 provides a novel strategy for pancreatic cancer therapy via overcoming radioresistance.
Collapse
Affiliation(s)
- Xiaoqing Wu
- Departments of Molecular Biosciences and Radiation Oncology, University of Kansas, Lawrence, KS, USA.,Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA.,School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Wenhua Tang
- Departments of Molecular Biosciences and Radiation Oncology, University of Kansas, Lawrence, KS, USA.,Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Rebecca T Marquez
- Departments of Molecular Biosciences and Radiation Oncology, University of Kansas, Lawrence, KS, USA
| | - Ke Li
- Departments of Molecular Biosciences and Radiation Oncology, University of Kansas, Lawrence, KS, USA
| | - Chad A Highfill
- Departments of Molecular Biosciences and Radiation Oncology, University of Kansas, Lawrence, KS, USA
| | - Fengtian He
- Departments of Molecular Biosciences and Radiation Oncology, University of Kansas, Lawrence, KS, USA.,Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, China
| | - Jiqin Lian
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, China
| | - Jiayuh Lin
- Department of Pediatrics, College of Medicine, Ohio State University, Columbus, OH, USA
| | - James R Fuchs
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Ohio State University, Columbus, OH, USA
| | - Min Ji
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Ling Li
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Cell Biology and Cell Engineering Research Centre, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shanxi, China
| | - Liang Xu
- Departments of Molecular Biosciences and Radiation Oncology, University of Kansas, Lawrence, KS, USA.,Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
183
|
Zhang ZL, Jiang QC, Wang SR. Schisandrin A reverses doxorubicin-resistant human breast cancer cell line by the inhibition of P65 and Stat3 phosphorylation. Breast Cancer 2017; 25:233-242. [PMID: 29181822 DOI: 10.1007/s12282-017-0821-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 11/15/2017] [Indexed: 01/07/2023]
Abstract
BACKGROUND Multidrug resistance (MDR) in breast cancer therapy occurs frequently. Thus, anti-MDR agents from natural products or synthetic compounds were tested extensively. We have also explored the reverse effect and mechanism of Schisandrin A (Sch A), a natural product, on MCF-7 breast cancer doxorubicin (DOX)-resistant subline MCF-7/DOX. METHODS MTT assay was performed to measure the viability of MCF-7 cells to assess the reverse effect of Sch A. Western blot analysis was used to study the protein levels. Laser scanning confocal microscopy was performed to detect the intercellular DOX and Rhodamine 123 accumulation. The qRT-PCR was used to analysis the target gene expression. Dual-luciferase reporter assay was performed to test the transcriptional activity of P-glycoprotein (P-gp). RESULTS Sch A, at the concentration of 20 µM, showed selective reverse effect (better than the positive control, verapamil at 5 µM) on MCF-7/DOX cell line but not on BEL-7402/DOX, Hep G2/DOX, and K-562/DOX cells. In addition, Sch A enhanced DOX-induced cleavage of Caspase-9 and PARP levels by increasing intracellular DOX accumulation and inhibiting P-gp function. Furthermore, Sch A selectively suppressed P-gp at gene and protein levels in MCF-7/DOX cells which express high level of MDR1 but not MRP1, MRP3, or BCRP. Besides, Sch A showed inhibitory effect on P-gp transcriptional activity. Sch A significantly reduced p-IκB-α (Ser32) and p-Stat3 (Tyr705) levels which mediate P-gp expression. In addition, Stat3 knockdown enhanced the reverse effect of siP65. The combined effect of siStat3 and siP65 was better than Sch A single treatment in MCF-7/DOX cells. CONCLUSION Sch A specifically reverses P-gp-mediated DOX resistance in MCF-7/DOX cells by blocking P-gp, NF-κB, and Stat3 signaling. Inhibition of P65 and Stat3 shows potent anti-MDR effect on MCF-7/DOX cells.
Collapse
Affiliation(s)
- Zong-Lin Zhang
- Department of Pharmacy, Linyi People's Hospital, Linyi, Shandong, China
| | - Qing-Cheng Jiang
- Department of Pharmacy, First People's Hospital of Tancheng County, Tancheng, Shandong, China
| | - Su-Rong Wang
- Department of Gynecology and Obstetrics, Linyi People's Hospital, 27# Jie fang lu dong duan, Linyi, Shandong, China.
| |
Collapse
|
184
|
Kanna R, Choudhary G, Ramachandra N, Steidl U, Verma A, Shastri A. STAT3 inhibition as a therapeutic strategy for leukemia. Leuk Lymphoma 2017; 59:2068-2074. [PMID: 29164994 DOI: 10.1080/10428194.2017.1397668] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Leukemia is characterized by selective overgrowth of malignant hematopoietic stem cells (HSC's) that interfere with HSC differentiation. Cytoreductive chemotherapy can kill rapidly dividing cancerous cells but cannot eradicate the malignant HSC pool leading to relapses. Leukemic stem cells have several dysregulated pathways and the Janus kinases (JAKs) and signal transducer and activator of transcription (STAT) pathway are prominent among them. STAT3 is an important transcription factor that regulates cell growth, proliferation, and inhibits apoptosis. High STAT3 expression in leukemia has been associated with an increased risk for relapse and decreased overall survival. Multiple strategies for interfering with STAT3 activity in leukemic cells include inhibition of STAT3 phosphorylation, interfering with STAT3 interactions, preventing nuclear transfer, inhibiting transcription and causing interference in STAT: DNA binding. A better understanding of key interactions and upstream mediators of STAT3 activity will help facilitate the development of effective cancer therapies and may result in durable remissions.
Collapse
Affiliation(s)
- Rubashruti Kanna
- a Division of Hematologic Malignancies, Department of Oncology, Albert Einstein College of Medicine , Bronx , NY , USA
| | - Gaurav Choudhary
- a Division of Hematologic Malignancies, Department of Oncology, Albert Einstein College of Medicine , Bronx , NY , USA
| | - Nandini Ramachandra
- a Division of Hematologic Malignancies, Department of Oncology, Albert Einstein College of Medicine , Bronx , NY , USA
| | - Ulrich Steidl
- a Division of Hematologic Malignancies, Department of Oncology, Albert Einstein College of Medicine , Bronx , NY , USA
| | - Amit Verma
- a Division of Hematologic Malignancies, Department of Oncology, Albert Einstein College of Medicine , Bronx , NY , USA
| | - Aditi Shastri
- a Division of Hematologic Malignancies, Department of Oncology, Albert Einstein College of Medicine , Bronx , NY , USA
| |
Collapse
|
185
|
Migrating into the Tumor: a Roadmap for T Cells. Trends Cancer 2017; 3:797-808. [DOI: 10.1016/j.trecan.2017.09.006] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/20/2017] [Accepted: 09/21/2017] [Indexed: 12/17/2022]
|
186
|
A pumpkin polysaccharide induces apoptosis by inhibiting the JAK2/STAT3 pathway in human hepatoma HepG2 cells. Int J Biol Macromol 2017. [DOI: 10.1016/j.ijbiomac.2017.06.078] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
187
|
High Content Imaging Assays for IL-6-Induced STAT3 Pathway Activation in Head and Neck Cancer Cell Lines. Methods Mol Biol 2017; 1683:229-244. [PMID: 29082496 DOI: 10.1007/978-1-4939-7357-6_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
In the canonical STAT3 signaling pathway, IL-6 receptor engagement leads to the recruitment of latent STAT3 to the activated IL-6 complex and the associated Janus kinase (JAK) phosphorylates STAT3 at Y705. pSTAT3-Y705 dimers traffic into the nucleus and bind to specific DNA response elements in the promoters of target genes to regulate their transcription. However, IL-6 receptor activation induces the phosphorylation of both the Y705 and S727 residues of STAT3, and S727 phosphorylation is required to achieve maximal STAT3 transcriptional activity. STAT3 continuously shuttles between the nucleus and cytoplasm and maintains a prominent nuclear presence that is independent of Y705 phosphorylation. The constitutive nuclear entry of un-phosphorylated STAT3 (U-STAT3) drives expression of a second round of genes by a mechanism distinct from that used by pSTAT3-Y705 dimers. The abnormally elevated levels of U-STAT3 produced by the constitutive activation of pSTAT3-Y705 observed in many tumors drive the expression of an additional set of pSTAT3-independent genes that contribute to tumorigenesis. In this chapter, we describe the HCS assay methods to measure IL-6-induced STAT3 signaling pathway activation in head and neck tumor cell lines as revealed by the expression and subcellular distribution of pSTAT3-Y705, pSTAT3-S727, and U-STAT3. Only the larger dynamic range provided by the pSTAT3-Y705 antibody would be robust and reproducible enough for screening.
Collapse
|
188
|
Galadari S, Rahman A, Pallichankandy S, Thayyullathil F. Molecular targets and anticancer potential of sanguinarine-a benzophenanthridine alkaloid. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 34:143-153. [PMID: 28899497 DOI: 10.1016/j.phymed.2017.08.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 07/06/2017] [Accepted: 08/06/2017] [Indexed: 05/03/2023]
Abstract
BACKGROUND Cancer is an enormous global health burden, and should be effectively addressed with better therapeutic strategies. Currently, over 60% of the clinically approved anticancer agents are either directly isolated from natural sources or are modified from natural lead molecules. Sanguinarine (SNG), a quaternary benzophenanthridine alkaloid has gained increasing attention in recent years as a potential anticancer agent. PURPOSE There is a large untapped source of phytochemical-based anticancer agents remaining to be explored. This review article aims to recapitulate different anticancer properties of SNG, and describes some of the molecular targets involved in exerting its effect. It also depicts the pharmacokinetic and toxicological properties of SNG, two parameters important in determining the druggability of a molecule. METHODS Numerous in vivo and in vitro published studies have signified the anticancer properties of SNG. In order to collate and decipher these properties, an extensive literature search was conducted in PubMed, ScienceDirect, and Scopus using keywords followed by the evaluation of the relevant articles where the relevant reports are integrated and analyzed. RESULTS Apart from inducing cell death, SNG inhibits pro-tumorigenic processes such as invasion, angiogenesis, and metastasis in different cancers. Moreover, SNG has been shown to synergistically enhance the sensitivity of several chemotherapeutic agents and is effective against a variety of multi-drug resistant cancers.
Collapse
Affiliation(s)
- Sehamuddin Galadari
- Cell Death Signaling Laboratory, Division of Science (Biology), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, UAE.
| | - Anees Rahman
- Cell Death Signaling Laboratory, Division of Science (Biology), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, UAE.
| | - Siraj Pallichankandy
- Cell Death Signaling Laboratory, Division of Science (Biology), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, UAE.
| | - Faisal Thayyullathil
- Cell Death Signaling Laboratory, Division of Science (Biology), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, UAE.
| |
Collapse
|
189
|
Lamichhane P, Karyampudi L, Shreeder B, Krempski J, Bahr D, Daum J, Kalli KR, Goode EL, Block MS, Cannon MJ, Knutson KL. IL10 Release upon PD-1 Blockade Sustains Immunosuppression in Ovarian Cancer. Cancer Res 2017; 77:6667-6678. [PMID: 28993412 DOI: 10.1158/0008-5472.can-17-0740] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/24/2017] [Accepted: 09/27/2017] [Indexed: 12/21/2022]
Abstract
Ligation of programmed cell death-1 (PD-1) in the tumor microenvironment is known to inhibit effective adaptive antitumor immunity. Blockade of PD-1 in humans has resulted in impressive, durable regression responses in select tumor types. However, durable responses have been elusive in ovarian cancer patients. PD-1 was recently shown to be expressed on and thereby impair the functions of tumor-infiltrating murine and human myeloid dendritic cells (TIDC) in ovarian cancer. In the present work, we characterize the regulation of PD-1 expression and the effects of PD-1 blockade on TIDC. Treatment of TIDC and bone marrow-derived dendritic cells (DC) with IL10 led to increased PD-1 expression. Both groups of DCs also responded to PD-1 blockade by increasing production of IL10. Similarly, treatment of ovarian tumor-bearing mice with PD-1 blocking antibody resulted in an increase in IL10 levels in both serum and ascites. While PD-1 blockade or IL10 neutralization as monotherapies were inefficient, combination of these two led to improved survival and delayed tumor growth; this was accompanied by augmented antitumor T- and B-cell responses and decreased infiltration of immunosuppressive MDSC. Taken together, our findings implicate compensatory release of IL10 as one of the adaptive resistance mechanisms that undermine the efficacy of anti-PD-1 (or anti-PD-L1) monotherapies and prompt further studies aimed at identifying such resistance mechanisms. Cancer Res; 77(23); 6667-78. ©2017 AACR.
Collapse
Affiliation(s)
- Purushottam Lamichhane
- Department of Immunology, Mayo Clinic Rochester, Minnesota.,Department of Immunology, Mayo Clinic Florida, Jacksonville, Florida.,The Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, Florida
| | - Lavakumar Karyampudi
- Department of Immunology, Mayo Clinic Rochester, Minnesota.,The Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, Florida
| | - Barath Shreeder
- Department of Immunology, Mayo Clinic Florida, Jacksonville, Florida.,The Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, Florida
| | - James Krempski
- Department of Immunology, Mayo Clinic Rochester, Minnesota
| | - Deborah Bahr
- Department of Immunology, Mayo Clinic Florida, Jacksonville, Florida
| | - Joshua Daum
- Department of Immunology, Mayo Clinic Florida, Jacksonville, Florida
| | | | - Ellen L Goode
- Department of Health Sciences Research, Mayo Clinic Rochester, Minnesota
| | | | - Martin J Cannon
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic Rochester, Minnesota. .,Department of Immunology, Mayo Clinic Florida, Jacksonville, Florida.,The Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, Florida
| |
Collapse
|
190
|
Effects of Thymoquinone on IL-6 Gene Expression and Some Cellular Signaling Pathways in Prostate Cancer PC3 Cells. Jundishapur J Nat Pharm Prod 2017. [DOI: 10.5812/jjnpp.63753] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
191
|
A Critical Review on the Effect of Docosahexaenoic Acid (DHA) on Cancer Cell Cycle Progression. Int J Mol Sci 2017; 18:ijms18081784. [PMID: 28817068 PMCID: PMC5578173 DOI: 10.3390/ijms18081784] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 08/14/2017] [Accepted: 08/16/2017] [Indexed: 11/17/2022] Open
Abstract
Globally, there were 14.1 million new cancer diagnoses and 8.2 million cancer deaths in 2012. For many cancers, conventional therapies are limited in their successes and an improved understanding of disease progression is needed in conjunction with exploration of alternative therapies. The long chain polyunsaturated fatty acid, docosahexaenoic acid (DHA), has been shown to enhance many cellular responses that reduce cancer cell viability and decrease proliferation both in vitro and in vivo. A small number of studies suggest that DHA improves chemotherapy outcomes in cancer patients. It is readily incorporated into cancer cell membranes and, as a result there has been considerable research regarding cell membrane initiated events. For example, DHA has been shown to mediate the induction of apoptosis/reduction of proliferation in vitro and in vivo. However, there is limited research into the effect of DHA on cell cycle regulation in cancer cells and the mechanism(s) by which DHA acts are not fully understood. The purpose of the current review is to provide a critical examination of the literature investigating the ability of DHA to stall progression during different cell cycle phases in cancer cells, as well as the consequences that these changes may have on tumour growth, independently and in conjunction with chemotherapy.
Collapse
|
192
|
Zulkifli AA, Tan FH, Putoczki TL, Stylli SS, Luwor RB. STAT3 signaling mediates tumour resistance to EGFR targeted therapeutics. Mol Cell Endocrinol 2017; 451:15-23. [PMID: 28088467 DOI: 10.1016/j.mce.2017.01.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 01/09/2017] [Indexed: 01/20/2023]
Abstract
Several EGFR inhibitors are currently undergoing clinical assessment or are approved for the clinical management of patients with varying tumour types. However, treatment often results in a lack of response in many patients. The majority of patients that initially respond eventually present with tumours that display acquired resistance to the original therapy. A large number of receptor tyrosine and intracellular kinases have been implicated in driving signaling that mediates this tumour resistance to anti-EGFR targeted therapy, and in a few cases these discoveries have led to overall changes in prospective tumour screening and clinical practice (K-RAS in mCRC and EGFR T790M in NSCLC). In this mini-review, we specifically focus on the role of the STAT3 signaling axis in providing both intrinsic and acquired resistance to inhibitors of the EGFR. We also focus on STAT3 pathway targeting in an attempt to overcome resistance to anti-EGFR therapeutics.
Collapse
Affiliation(s)
- Ahmad A Zulkifli
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Fiona H Tan
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Tracy L Putoczki
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050, Australia
| | - Stanley S Stylli
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Rodney B Luwor
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
| |
Collapse
|
193
|
Binder S, Hösler N, Riedel D, Zipfel I, Buschmann T, Kämpf C, Reiche K, Burger R, Gramatzki M, Hackermüller J, Stadler PF, Horn F. STAT3-induced long noncoding RNAs in multiple myeloma cells display different properties in cancer. Sci Rep 2017; 7:7976. [PMID: 28801664 PMCID: PMC5554185 DOI: 10.1038/s41598-017-08348-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 07/10/2017] [Indexed: 12/31/2022] Open
Abstract
Interleukin-6 (IL-6)-activated Signal Transducer and Activator of Transcription 3 (STAT3) facilitates survival in the multiple myeloma cell line INA-6 and therefore represents an oncogenic key player. However, the biological mechanisms are still not fully understood. In previous studies we identified microRNA-21 as a STAT3 target gene with strong anti-apoptotic potential, suggesting that noncoding RNAs have an impact on the pathogenesis of human multiple myeloma. Here, we describe five long noncoding RNAs (lncRNAs) induced by IL-6-activated STAT3, which we named STAiRs. While STAiRs 1, 2 and 6 remain unprocessed in the nucleus and show myeloma-specific expression, STAiRs 15 and 18 are spliced and broadly expressed. Especially STAiR2 and STAiR18 are promising candidates. STAiR2 originates from the first intron of a tumor suppressor gene. Our data support a mutually exclusive expression of either STAiR2 or the functional tumor suppressor in INA-6 cells and thus a contribution of STAiR2 to tumorigenesis. Furthermore, STAiR18 was shown to be overexpressed in every tested tumor entity, indicating its global role in tumor pathogenesis. Taken together, our study reveals a number of STAT3-induced lncRNAs suggesting that the interplay between the coding and noncoding worlds represents a fundamental principle of STAT3-driven cancer development in multiple myeloma and beyond.
Collapse
Affiliation(s)
- Stefanie Binder
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany.
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.
- LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany.
| | - Nadine Hösler
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Diana Riedel
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
| | - Ivonne Zipfel
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
| | - Tilo Buschmann
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
- The RIBOLUTION Consortium, Leipzig, Germany
| | - Christoph Kämpf
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
- Young Investigators Group Bioinformatics and Transcriptomics, Department Proteomics, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
- Bioinformatics Group, Department of Computer Science, University of Leipzig, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Kristin Reiche
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
- The RIBOLUTION Consortium, Leipzig, Germany
- Young Investigators Group Bioinformatics and Transcriptomics, Department Proteomics, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Renate Burger
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine 2, Christian-Albrechts-University, Kiel, Germany
| | - Martin Gramatzki
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine 2, Christian-Albrechts-University, Kiel, Germany
| | - Jörg Hackermüller
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
- Young Investigators Group Bioinformatics and Transcriptomics, Department Proteomics, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
- Department of Computer Science, University of Leipzig, Leipzig, Germany
| | - Peter F Stadler
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
- Bioinformatics Group, Department of Computer Science, University of Leipzig, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany
- German Centre for Integrative Biodiversity Research - iDiv, Halle-Jena-Leipzig, Germany
- Max Planck Institute for Mathematics in the Sciences, Leipzig, Germany
- Department of Theoretical Chemistry, University of Vienna, Vienna, Austria
- Center for RNA in Technology and Health, University of Copenhagen, Copenhagen, Denmark
- Santa Fe Institute, Santa Fe, USA
| | - Friedemann Horn
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
- The RIBOLUTION Consortium, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| |
Collapse
|
194
|
Metelev V, Zhang S, Zheng S, Kumar AT, Bogdanov A. Fluorocarbons Enhance Intracellular Delivery of Short STAT3-sensors and Enable Specific Imaging. Am J Cancer Res 2017; 7:3354-3368. [PMID: 28900515 PMCID: PMC5595137 DOI: 10.7150/thno.19704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 07/12/2017] [Indexed: 02/06/2023] Open
Abstract
Short oligonucleotide sequences are now being widely investigated for their potential therapeutic properties. The modification of oligonucleotide termini with short fluorinated residues is capable of drastically altering their behavior in complex in vitro and in vivo systems, and thus may serve to greatly enhance their therapeutic potential. The main goals of our work were to explore: 1) how modification of STAT3 transcription factor-binding oligodeoxynucleotide (ODN) duplexes (ODND) with one or two short fluorocarbon (FC)-based residues would change their properties in vitro and in vivo, and if so, how this would affect their intracellular uptake by cancer cells, and 2) the ability of such modified ODND to form non-covalent complexes with FC-modified carrier macromolecule. The latter has an inherent advantage of producing a 19F-specific magnetic resonance (MR) imaging signature. Thus, we also tested the ability of such copolymers to generate 19F-MR signals. Materials and Methods. Fluorinated nucleic acid residues were incorporated into ODN by using automated synthesis or via activated esters on ODN 5'-ends. To quantify ODND uptake by the cells and to track their stability, we covalently labeled ODN with fluorophores using internucleoside linker technology; the FC-modified carrier was synthesized by acylation of pegylated polylysine graft copolymer with perfluoroundecanoic acid (M5-gPLL-PFUDA). Results. ODN with a single FC group exhibited a tendency to form duplexes with higher melting points and with increased stability against degradation when compared to control non-modified ODNs. ODND carrying fluorinated residues showed complex formation with M5-gPLL-PFUDA as predicted by molecular dynamics simulations. Moreover, FC groups modulated the specificity of ODND binding to the STAT3 target. Finally, FC modification resulted in greater cell uptake (2 to 4 fold higher) when compared to the uptake of non-modified ODND as determined by quantitative confocal fluorescence imaging of A431 and INS-1 cells. Conclusion. ODND modification with FC residues enables fine-tuning of protein binding specificity to double-strand binding motifs and results in an increased internalization by A431 and INS-1 cells in culture. Our results show that modification of ODN termini with FC residues is both a feasible and powerful strategy for developing more efficient nucleic acid-based therapies with the added benefit of allowing for non-invasive MR imaging of ODND therapeutic targeting and response.
Collapse
|
195
|
Zhao X, Huang L, Xu W, Chen X, Shen Y, Zeng W, Chen X. Physapubescin B inhibits tumorgenesis and circumvents taxol resistance of ovarian cancer cells through STAT3 signaling. Oncotarget 2017; 8:70130-70141. [PMID: 29050266 PMCID: PMC5642541 DOI: 10.18632/oncotarget.19593] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 06/28/2017] [Indexed: 01/05/2023] Open
Abstract
Ovarian cancer is the most lethal gynaecological malignancy. Recurrence and subsequent resistance to chemotherapy have become major obstacles to treating these diseases. In the present study, we showed that a natural withanolide isolated from the plant Physalis pubescens L. (Solanaceae), Physapubescin B, exhibited potent anti-tumor activity against ovarian cancer cells. Physapubescin B promoted apoptosis, induced cell-cycle arrest and inhibited invasion of ES-2 and A2780 cells. Physapubescin B treatment also resulted in suppression of the transcriptional activity of STAT3, an oncogenic transcription factor activated in many human malignancies including ovarian cancer, through disturbing the dimerization of STAT3, and thereby inhibited the nuclear translocation of Tyr705/Ser727-phosphorylated STAT3. The IL-6-stimulated activation of STAT3 and its downstream genes Cyclin D1, survivin, and Bcl-xL was also repressed by Physapubescin B. Furthermore, Physapubescin B sensitizes A2780 cells to taxol-induced cell growth inhibition in vitro. These findings strongly suggest that Physapubescin B has potential antitumor activity and may circumvent taxol resistance in human ovarian cancer cells through inhibition of aberrant activation of STAT3.
Collapse
Affiliation(s)
- Xiaofeng Zhao
- Department of Gynecology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang Province, China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Lu Huang
- Department of Gynecology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang Province, China
| | - Wanwan Xu
- Department of Gynecology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang Province, China.,Bengbu Medical College, Bengbu, Anhui Province, China
| | - Xiaoyan Chen
- Department of Gynecology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang Province, China
| | - Yan Shen
- Department of Gynecology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang Province, China
| | - Wenjie Zeng
- Department of Gynecology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang Province, China
| | - Xiao Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
196
|
Tsurumaki H, Katano H, Sato K, Imai R, Niino S, Hirabayashi Y, Ichikawa S. WP1066, a small molecule inhibitor of the JAK/STAT3 pathway, inhibits ceramide glucosyltransferase activity. Biochem Biophys Res Commun 2017; 491:265-270. [PMID: 28739255 DOI: 10.1016/j.bbrc.2017.07.115] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 07/20/2017] [Indexed: 02/05/2023]
Abstract
WP1066 is a well-known inhibitor of the JAK/STAT3 signaling pathway. By a screen of known small molecule inhibitors of various enzymes and protein factors, we identified WP1066 as a ceramide glucosyltransferase inhibitor. Ceramide glucosyltransferase catalyzes the first glycosylation step during glycosphingolipid synthesis. We found that WP1066 inhibited the activity of ceramide glucosyltransferase with an IC50 of 7.2 μM, and that its action was independent of JAK/STAT3 pathway blockade. Moreover, the modes of inhibition of ceramide glucosyltransferase were uncompetitive with respect to both C6-NBD-cermide and UDP-glucose.
Collapse
Affiliation(s)
- Hirotaka Tsurumaki
- Laboratory for Animal Cell Engineering, Niigata University of Pharmacy and Applied Life Sciences (NUPALS), 265-1 Higashijima, Akiha-ku, Niigata-shi, Niigata 956-8603, Japan
| | - Hikaru Katano
- Laboratory for Animal Cell Engineering, Niigata University of Pharmacy and Applied Life Sciences (NUPALS), 265-1 Higashijima, Akiha-ku, Niigata-shi, Niigata 956-8603, Japan
| | - Kousuke Sato
- Laboratory for Animal Cell Engineering, Niigata University of Pharmacy and Applied Life Sciences (NUPALS), 265-1 Higashijima, Akiha-ku, Niigata-shi, Niigata 956-8603, Japan
| | - Ryou Imai
- Laboratory for Animal Cell Engineering, Niigata University of Pharmacy and Applied Life Sciences (NUPALS), 265-1 Higashijima, Akiha-ku, Niigata-shi, Niigata 956-8603, Japan
| | - Satomi Niino
- Laboratory for Animal Cell Engineering, Niigata University of Pharmacy and Applied Life Sciences (NUPALS), 265-1 Higashijima, Akiha-ku, Niigata-shi, Niigata 956-8603, Japan
| | - Yoshio Hirabayashi
- Laboratory for Molecular Membrane Neuroscience, Brain Science Institute (BSI), The Institute of Physical and Chemical Research (RIKEN), 2-1 Hirosawa, Wako-shi, Saitama 351-01, Japan
| | - Shinichi Ichikawa
- Laboratory for Animal Cell Engineering, Niigata University of Pharmacy and Applied Life Sciences (NUPALS), 265-1 Higashijima, Akiha-ku, Niigata-shi, Niigata 956-8603, Japan.
| |
Collapse
|
197
|
Chen C, Wang X, Fang J, Xue J, Xiong X, Huang Y, Hu J, Ling K. EGFR-induced phosphorylation of type Iγ phosphatidylinositol phosphate kinase promotes pancreatic cancer progression. Oncotarget 2017; 8:42621-42637. [PMID: 28388589 PMCID: PMC5522093 DOI: 10.18632/oncotarget.16730] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 01/24/2017] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer is one of the deadliest malignancies and effective treatment has always been lacking. In current study, we investigated how the type Iγ phosphatidylinositol phosphate kinase (PIPKIγ) participates in the progression of pancreatic ductal adenocarcinoma (PDAC) for novel therapeutic potentials against this lethal disease. We found that PIPKIγ is up-regulated in all tested PDAC cell lines. The growth factor (including EGFR)-induced tyrosine phosphorylation of PIPKIγ is significantly elevated in in situ and metastatic PDAC tissues. Loss of PIPKIγ inhibits the aggressiveness of PDAC cells by restraining the activities of AKT and STAT3, as well as MT1-MMP expression. Therefore when planted into the pancreas of nude mice, PIPKIγ-depleted PDAC cells exhibits substantially repressed tumor growth and metastasis comparing to control PDAC cells. Results from further studies showed that the phosphorylation-deficient PIPKIγ mutant, unlike its wild-type counterpart, cannot rescue PDAC progression inhibited by PIPKIγ depletion. These findings indicate that PIPKIγ, functioning downstream of EGFR signaling, is critical to the progression of PDAC, and suggest that PIPKIγ is potentially a valuable therapeutic target for PDAC treatment.
Collapse
Affiliation(s)
- Chunhua Chen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- These authors have contributed equally to this work
| | - Xiangling Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- These authors have contributed equally to this work
| | - Juemin Fang
- Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- These authors have contributed equally to this work
| | - Junli Xue
- Shanghai East Hospital, Tongji University, Shanghai, China
| | - Xunhao Xiong
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Yan Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jinghua Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kun Ling
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
198
|
Wu Z, Guo L, Ge J, Zhang Z, Wei H, Zhou Q. Two serine residues of non-metastasis protein 23-H1 are critical in inhibiting signal transducer and activator of transcription 3 activity in human lung cancer cells. Oncol Lett 2017; 14:2475-2482. [PMID: 28781685 DOI: 10.3892/ol.2017.6363] [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: 02/16/2016] [Accepted: 02/13/2017] [Indexed: 02/05/2023] Open
Abstract
Constitutive activation of signal transducer and activator of transcription 3 (STAT3) in numerous cancers, including lung cancer, is one of the major mechanisms of tumor progression and metastasis. The authors previously reported that the metastasis suppressor non-metastasis protein 23-H1 (Nm23-H1) negatively regulates STAT3 activity by inhibiting its phosphorylation on Tyr705. Nm23-H1 is a multifunction protein that has three different kinase activities. By transfecting the five mutants that inactivated three different kinase activities respectively into Nm23-H1 deficient lung cancer cell lines, it was identified that Nm23-H1S44A (Ser44 to Ala) and Nm23-H1S120G (Ser120 to Gly) mutant forms were unable to suppress STAT3 phosphorylation on Tyr705, resulting in increased expression of fibronectin and matrix metalloproteinase-9. Notably, protein inhibitor of activated STAT3 was also involved in Nm23-H1S44A- and Nm23-H1S120G-mediated suppression of STAT3 phosphorylation. The present results indicated that Ser44 and Ser120 sites of Nm23-H1 may be responsible for its biological suppressive effects of STAT3 and tumor metastasis, which may contribute to illuminate the metastasis suppression function of Nm23-H1 in lung cancer.
Collapse
Affiliation(s)
- Zhihao Wu
- Department of Medical Biology, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Lili Guo
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Center and Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Hexi, Tianjin 300060, P.R. China
| | - Jiangnan Ge
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Center and Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Zhijian Zhang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Center and Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Huijun Wei
- Department of Central Laboratory, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Qinghua Zhou
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Center and Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Sichuan Lung Cancer Institute, Sichuan Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, P.R. China
| |
Collapse
|
199
|
Li T, Fu X, Tse AKW, Guo H, Lee KW, Liu B, Su T, Wang X, Yu Z. Inhibiting STAT3 signaling is involved in the anti-melanoma effects of a herbal formula comprising Sophorae Flos and Lonicerae Japonicae Flos. Sci Rep 2017; 7:3097. [PMID: 28596565 PMCID: PMC5465088 DOI: 10.1038/s41598-017-03351-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/26/2017] [Indexed: 12/14/2022] Open
Abstract
A herbal formula (SL) comprising Sophorae Flos and Lonicerae Japonicae Flos was traditionally used to treat melanoma. Constitutively active signal transducer and activator of transcription 3 (STAT3) has been proposed as a therapeutic target in melanoma. Here we investigated whether an ethanolic extract of SL (SLE) exerted anti-melanoma activities by inhibiting STAT3 signaling. B16F10 allograft model, A375 and B16F10 cells were employed to assess the in vivo and in vitro anti-melanoma activities of SLE. A375 cells stably expressing STAT3C, a constitutively active STAT3 mutant, were used to determine the role of STAT3 signaling in SLE’s anti-melanoma effects. Intragastric administration of SLE (1.2 g/kg) potently inhibited melanoma growth in mice and inhibited STAT3 phosphorylation in the tumors. In cultured cells, SLE dramatically reduced cell viability, induced apoptosis, suppressed migration and invasion, and restrained STAT3 activation and nuclear localization. STAT3C overexpression in A375 cells diminished SLE’s effects on cell viability, apoptosis and invasion. Collectively, SLE exerted potent anti-melanoma effects partially by inhibiting STAT3 signaling. This study provides pharmacological justification for the traditional use of this formula in treating melanoma, and suggests that SLE has the potential to be developed as a modern alternative and/or complimentary agent for melanoma treatment and prevention.
Collapse
Affiliation(s)
- Ting Li
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Xiuqiong Fu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Anfernee Kai-Wing Tse
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Hui Guo
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Kin Wah Lee
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Bin Liu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Tao Su
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Xueyu Wang
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Zhiling Yu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China. .,Research and Development Centre for Natural Health Products, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.
| |
Collapse
|
200
|
Cao S, Chen C, Xue J, Huang Y, Yang X, Ling K. Silencing of type Iγ phosphatidylinositol phosphate kinase suppresses ovarian cancer cell proliferation, migration and invasion. Oncol Rep 2017; 38:253-262. [PMID: 28560454 PMCID: PMC5492676 DOI: 10.3892/or.2017.5670] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 05/05/2017] [Indexed: 12/18/2022] Open
Abstract
Metastasis is the major cause of death in ovarian cancer patients. Given that the molecular mechanism underlying metastasis formation is critical for improving therapeutic development and clinical treatment, it must be fully understood. Recent studies have revealed that lipid kinase type Iγ phosphatidylinositol phosphate kinase (PIPKIγ) participates in the metastasis of breast cancer and colon cancer by regulating cell migration and invasion. However, its role in the progression of ovarian cancer is unclear. Here we showed that PIPKIγ expression is upregulated in multiple epithelial ovarian cancer cell lines. Silencing of PIPKIγ impaired PI3K/AKT signaling and inhibited the aggressive behaviors of epithelial ovarian cancer cells, including proliferation, migration and invasion. Moreover, we found that PIPKIγ was required for the activation of signal transducer and activator of transcription 3 (STAT3) in epithelial ovarian cancer cells, indicating that STAT3 may also be engaged in the PIPKIγ-dependent aggressiveness of epithelial ovarian cancer cells. Our results, for the first time, identified PIPKIγ as a novel regulator in epithelial ovarian cancer cells that promotes cell proliferation, migration and invasion by activating multiple signaling pathways. Therefore, we propose that PIPKIγ could potentially be a therapeutic target for the early detection and treatment of epithelial ovarian cancer. Further studies employing in vivo models are necessary to test this possibility.
Collapse
Affiliation(s)
- Siyu Cao
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of the Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Chunhua Chen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55901, USA
| | - Junli Xue
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Yan Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55901, USA
| | - Xiaofeng Yang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of the Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Kun Ling
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55901, USA
| |
Collapse
|