1
|
Aloui M, El fadili M, Mujwar S, Er-rahmani S, Abuelizz HA, Er-rajy M, Zarougui S, Elhallaoui M. Design of novel potent selective survivin inhibitors using 2D-QSAR modeling, molecular docking, molecular dynamics, and ADMET properties of new MX-106 hydroxyquinoline scaffold derivatives. Heliyon 2024; 10:e38383. [PMID: 39397921 PMCID: PMC11467593 DOI: 10.1016/j.heliyon.2024.e38383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024] Open
Abstract
Given the critical role of survivin (BIRC5) in tumor cell regulation, developing novel inhibitors represents a promising approach for cancer therapy. This study details the design of innovative survivin inhibitors based on the hydroxyquinoline scaffold of our previously reported lead compound, MX-106. Our study identified nine compounds whose inhibitory activity is expected to be superior to that of the most active molecule in the series. These compounds demonstrated potent suppression of MDA-MB-435 breast cancer cell proliferation in vitro and exhibited enhanced metabolic stability compared to the series' most active member. To evaluate these derivatives as potential survivin inhibitors, we employed a multi-faceted approach combining 2D-QSAR methods, molecular docking, molecular dynamics, and ADMET property assessment. Our molecular modeling studies led to the design of nine novel compounds (Pred1-Pred9) predicted to exhibit potent survivin inhibitory activity based on MLR models. To assess their suitability as drug candidates, we recommend a thorough evaluation of their ADMET properties. These compounds hold promise as innovative anticancer agents targeting survivin, similar to the established MX-106.
Collapse
Affiliation(s)
- Mourad Aloui
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Mohamed El fadili
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Somdutt Mujwar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Sara Er-rahmani
- Dipartimento di Chimica, Università di Torino, 10125, Torino, Italy
| | - Hatem A. Abuelizz
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, PO Box 2457, Riyadh, 11451, Saudi Arabia
| | - Mohammed Er-rajy
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Sara Zarougui
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Menana Elhallaoui
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
2
|
Xu W, Hu J, Ma Z, Feng W, Gong W, Fu S, Chen X. Decreased BIRC5-206 promotes epithelial-mesenchymal transition in nasopharyngeal carcinoma through sponging miR-145-5p. Open Med (Wars) 2024; 19:20241007. [PMID: 39308922 PMCID: PMC11416051 DOI: 10.1515/med-2024-1007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/23/2024] [Accepted: 07/01/2024] [Indexed: 09/25/2024] Open
Abstract
Metastasis significantly contributes to the poor prognosis of advanced nasopharyngeal carcinoma (NPC). Our prior studies have demonstrated a decrease in BIRC5-206 expression in NPC, which promotes disease progression. However, the role of BIRC5-206 in the invasion and metastasis of NPC has not been fully elucidated. In this study, our objective was to explore the biological function and underlying mechanisms of BIRC5-206 in NPC. Additionally, we established an NPC mouse model of lung invasiveness using C666 cells to assess the impact of BIRC5-206 on NPC metastasis. Our results revealed that silencing BIRC5-206 inhibited apoptosis and enhanced the invasion of NPC cells, whereas its overexpression reversed these effects. Moreover, decreased BIRC5-206 expression significantly increased N-cadherin and Vimentin expression while reducing E-cadherin and occludin levels, both in vivo and in vitro. Additionally, silencing BIRC5-206 markedly augmented the formation of invasive foci in lung tissues. Rescue experiments further confirmed that decreased BIRC5-206 expression facilitates NPC metastasis via modulation of the miR-145-5p/CD40 signaling pathway. In summary, our study suggests that BIRC5-206 may serve as a potential prognostic biomarker and therapeutic target in the diagnosis and treatment of NPC.
Collapse
Affiliation(s)
- Weihua Xu
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Hainan Tropical Cancer Research Institute, Haikou, Hainan, 570312, China
| | - Junjie Hu
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Hainan Tropical Cancer Research Institute, Haikou, Hainan, 570312, China
| | - Zhichao Ma
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Hainan Tropical Cancer Research Institute, Haikou, Hainan, 570312, China
| | - Wanyi Feng
- Hainan Lvtou Medical Laboratory Center, Haikou, Hainan, 570206, China
- School of Life Sciences, Hainan University, Haikou, Hainan, 570228, China
| | - Wei Gong
- Hainan Lvtou Medical Laboratory Center, Haikou, Hainan, 570206, China
- School of Life Sciences, Hainan University, Haikou, Hainan, 570228, China
| | - Shengmiao Fu
- Central Laboratory, Hainan General Hospital, Hainan Hospital Affiliated to the Hainan Medical College, No. 19 Xiuhua Road, Xiuying District, Haikou, Hainan, 570311, China
- Hainan Lvtou Medical Laboratory Center, No. 16 Jinyu East Road, Longhua District, Haikou, Hainan, 570206, China
| | - Xinping Chen
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Hainan Tropical Cancer Research Institute, No. 6, Changbin West 4th Street, Xiuying District, Haikou, Hainan, 570312, China
| |
Collapse
|
3
|
Ebadi Sharafabad B, Abdoli A, Panahi M, Abdolmohammadi Khiav L, Jamur P, Abedi Jafari F, Dilmaghani A. Anti-tumor Effects of Cisplatin Synergist in Combined Treatment with Clostridium novyi-NT Spores Against Hypoxic Microenvironments in a Mouse Model of Cervical Cancer Caused by TC-1 Cell Line. Adv Pharm Bull 2023; 13:817-826. [PMID: 38022809 PMCID: PMC10676560 DOI: 10.34172/apb.2023.084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 04/26/2023] [Accepted: 05/17/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Despite the development of anti-human papillomavirus (HPV) vaccines, cervical cancer is still a common disease in women, especially in developing countries. The presence of a hypoxic microenvironment causes traditional treatments to fail. In this study, we presented a combined treatment method based on the chemotherapeutic agent cisplatin and Clostridium novyi-NT spores to treat normoxic and hypoxic areas of the tumor. Methods TC-1 Cell line capable of expressing HPV-16 E6/7 oncoproteins was subcutaneously transplanted into female 6-8 week old C57/BL6 mice. The tumor-bearing mice were randomly divided into four groups and treated with different methods after selecting a control group. Group 1: Control without treatment (0.1 mL sterile PBS intratumorally), Group: C. novyi-NT (107 C. novyi-NT). Group 3: Receives cisplatin intraperitoneally (10 mg/kg). Fourth group: Intratumoral administration of C. novyi-NT spores + intraperitoneal cisplatin. Western blot analysis was used to examine the effects of anti-hypoxia treatment and expression of hypoxia-inducible factor 1 (HIF-1) and vascular endothelial growth factor (VEGF) proteins. Results The results clearly showed that combined treatment based on C. novyi-NT and cisplatin significantly reduced the expression of HIF-1 alpha and VEGF proteins compared to cisplatin alone. At the same time, the amount of necrosis of tumor cells in the combined treatment increased significantly compared to the single treatment and the control. At the same time, the mitotic count decreased significantly. Conclusion Our research showed that developing a combined treatment method based on C. novyi-NT and cisplatin against HPV-positive cervical cancer could overcome the treatment limitations caused by the existence of hypoxic areas of the tumor.
Collapse
Affiliation(s)
- Behrouz Ebadi Sharafabad
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asghar Abdoli
- Department of Hepatitis and HIV, Pasteur Institute of Iran (IPI), Tehran, Iran
| | - Mohammad Panahi
- Department of Hepatitis and HIV, Pasteur Institute of Iran (IPI), Tehran, Iran
| | - Lida Abdolmohammadi Khiav
- Department of Anaerobic Vaccine Research and Production, Specialized Clostridia Research Laboratory, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj, Iran
| | - Parisa Jamur
- Department of Hepatitis and HIV, Pasteur Institute of Iran (IPI), Tehran, Iran
| | - Fatemeh Abedi Jafari
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Azita Dilmaghani
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Li Y, He W, Gao X, Lu X, Xie F, Um SW, Kang MW, Yang H, Shang Y, Wang Z, Fu J, Jia Y. Cullin7 induces docetaxel resistance by regulating the protein level of the antiapoptotic protein Survivin in lung adenocarcinoma cells. J Thorac Dis 2023; 15:5006-5019. [PMID: 37868891 PMCID: PMC10586960 DOI: 10.21037/jtd-23-1110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/08/2023] [Indexed: 10/24/2023]
Abstract
Background Lung adenocarcinoma (LUAD) is the most common subtype of non-small cell lung cancer (NSCLC). Chemotherapy resistance is the main cause of chemotherapy failure. Cullin7 (Cul7) is highly expressed in LUAD and is associated with poor prognosis. Moreover, Cul7 is abnormally overexpressed in docetaxel-resistant LUAD cells. Therefore, further exploration of the role and molecular mechanism of Cul7 in LUAD docetaxel resistance is necessary. Methods We established docetaxel-resistant cell lines (A549DTX and H358DTX cell lines) by exposing cells to gradually increasing concentrations of docetaxel. Cell (A549, A549DTX, H358, and H358DTX cell lines) sensitivity to docetaxel was determined via a 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymmethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS) assay. And then quantitative polymerase chain reaction (qPCR) and Western blotting were performed to measure the expression of Cul7 and Survivin in A549, A549DTX, H358, and H358DTX cell lines. Subsequently, we knocked down Cul7 in docetaxel-resistant cells and overexpressed Cul7 in parental cells via lentiviral transduction to further validate the correlation between Cul7 and docetaxel resistance, while exploring the molecular mechanism of docetaxel resistance it caused. Immunofluorescence and immunohistochemical (IHC) staining were also used to evaluate the expression and cellular localization of Cul7. To confirm the effect of Cul7 expression on cell apoptosis, we used flow cytometry to detect the apoptosis rate of A549 and A549DTX cells with the same drug concentration. Results Cul7 was highly expressed in A549DTX and H358DTX cells. However, when Cul7 expression was knocked down in A549DTX and H358DTX cells, cell sensitivity to docetaxel was significantly increased. In addition, we found that Cul7 was coexpressed with Survivin. Silencing Survivin reversed the docetaxel insensitivity caused by Cul7 overexpression. High expression of Cul7 and Survivin in docetaxel-resistant LUAD cells inhibited the intrinsic apoptosis pathway and promoted cell proliferation. Therefore, the Cul7/Survivin axis may play a role in inducing LUAD docetaxel chemoresistance. Conclusions Cul7 and Survivin were both highly expressed in docetaxel-resistant LUAD cells. Our results suggest that Cul7 may inhibit apoptosis and promote the proliferation of LUAD cells by increasing the Survivin protein level, which in turn contributes to docetaxel chemoresistance in LUAD.
Collapse
Affiliation(s)
- Yumiao Li
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Wenyi He
- College of Clinical Medicine, Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Xiangpeng Gao
- College of Clinical Medicine, Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Xiaomei Lu
- GZ Runsheng CytoMed Technology Co., Ltd., Guangzhou, China
| | - Fangni Xie
- Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
| | - Sang-Won Um
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Min-Woong Kang
- Department of Thoracic and Cardiovascular Surgery, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Hua Yang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Yanhong Shang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Zhiyu Wang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Jiejun Fu
- Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
| | - Youchao Jia
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| |
Collapse
|
5
|
Shoshan-Barmatz V, Arif T, Shteinfer-Kuzmine A. Apoptotic proteins with non-apoptotic activity: expression and function in cancer. Apoptosis 2023; 28:730-753. [PMID: 37014578 PMCID: PMC10071271 DOI: 10.1007/s10495-023-01835-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2023] [Indexed: 04/05/2023]
Abstract
Apoptosis is a process of programmed cell death in which a cell commits suicide while maintaining the integrity and architecture of the tissue as a whole. Apoptosis involves activation of one of two major pathways: the extrinsic pathway, where extracellular pro-apoptotic signals, transduced through plasma membrane death receptors, activate a caspase cascade leading to apoptosis. The second, the intrinsic apoptotic pathway, where damaged DNA, oxidative stress, or chemicals, induce the release of pro-apoptotic proteins from the mitochondria, leading to the activation of caspase-dependent and independent apoptosis. However, it has recently become apparent that proteins involved in apoptosis also exhibit non-cell death-related physiological functions that are related to the cell cycle, differentiation, metabolism, inflammation or immunity. Such non-conventional activities were predominantly reported in non-cancer cells although, recently, such a dual function for pro-apoptotic proteins has also been reported in cancers where they are overexpressed. Interestingly, some apoptotic proteins translocate to the nucleus in order to perform a non-apoptotic function. In this review, we summarize the unconventional roles of the apoptotic proteins from a functional perspective, while focusing on two mitochondrial proteins: VDAC1 and SMAC/Diablo. Despite having pro-apoptotic functions, these proteins are overexpressed in cancers and this apparent paradox and the associated pathophysiological implications will be discussed. We will also present possible mechanisms underlying the switch from apoptotic to non-apoptotic activities although a deeper investigation into the process awaits further study.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel.
- National Institute for Biotechnology in the Negev, Beer Sheva, Israel.
| | - Tasleem Arif
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | |
Collapse
|
6
|
Fanian M, Rafiei G, Alizadeh Zarei M, Takhshid MA. Targeted Overexpression of NDRG2 using Survivin Promoter Reduces Viability and Invasiveness of A549 Cell Line. Avicenna J Med Biotechnol 2023; 15:84-90. [PMID: 37034887 PMCID: PMC10073922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 01/24/2023] [Indexed: 04/11/2023] Open
Abstract
Background Anti-tumor effects of N-myc Downstream Regulated Gene2 (NDRG2) have been demonstrated in many tumors. In the present study, NDRG2 was specifically overexpressed in lung cancer cell line using Survivin Promoter (Sur-P). Then, the effects of NDRG2 overexpression on viability, apoptosis, migration, and invasion of A549 cells were evaluated. Methods Recombinant pAdenoVator-Sur-P-NDRG2-IRES-GFP plasmid harboring NDRG2 gene under transcriptional control of Sur-P and mock plasmid were constructed. A549 lung tumor cells and LX-2 cells (non-tumor cell line) were transfected with pAdenoVator-Sur-P-NDRG2-IRES-GFP, pAdenoVator-CMV-NDRG2-IRES-GFP, or mock plasmids. Tumor specificity of Sur-P was evaluated using fluorescent microscopy for GFP expression. The effects of NDRG2 overexpression on cell viability, apoptosis, and migration of A549 cells were measured using MTT, annexinV/7-AAD flow cytometry, and transwell migration assay, respectively. NDRG2 and matrix metalloproteinase-2 (MMP-2) expression were measured using real time-PCR. Results pAdenoVator-Sur-P-NDRG2-IRES-GFP transfection resulted in a huge GFP expression in A549 cells, but not in LX-2 cells. The results of real time-PCR analysis also showed that pAdenoVator-Sur-P-NDRG2-IRES-GFP transfection led to an abundant NDRG2 expression in A549 cells. NDRG2 overexpression decreased A549 cell viability through increasing cell apoptosis. In addition, migration, invasion, and MMP-2 expression decreased following NDRG2 overexpression in A549 cells. Conclusion The findings indicate that the targeted overexpression of NDRG2 using Sur-P can reduce the viability and invasiveness of A549 cells, suggesting possible benefits of this approach in lung cancer therapy.
Collapse
Affiliation(s)
- Maryam Fanian
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Gholamreza Rafiei
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Alizadeh Zarei
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Ali Takhshid
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Kyaw KZ, Byun WS, Shin YH, Huynh TH, Lee JY, Bae ES, Park HJ, Oh DC, Lee SK. Antitumor Activity of Piceamycin by Upregulation of N-Myc Downstream-Regulated Gene 1 in Human Colorectal Cancer Cells. JOURNAL OF NATURAL PRODUCTS 2022; 85:2817-2827. [PMID: 36458922 DOI: 10.1021/acs.jnatprod.2c00832] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Piceamycin (1), a macrocyclic lactam isolated from the silkworm's gut (Streptomyces sp. SD53 strain), reportedly possesses antibacterial activity. However, the potential anticancer activity and molecular processes underlying 1 have yet to be reported. Colorectal cancer (CRC) is high-risk cancer and accounts for 10% of all cancer cases worldwide. The high prevalence of resistance to radiation or chemotherapy means that patients with advanced CRC have a poor prognosis, with high recurrence and metastasis potential. Therefore, the present study investigated the antitumor effect and underlying mechanisms of 1 in CRC cells. The growth-inhibiting effect of 1 in CRC cells was correlated with the upregulation of a tumor suppressor, N-myc downstream-regulated gene 1 (NDRG1). Additionally, 1 induced G0/G1 cell cycle arrest and apoptosis and inhibited the migration of CRC cells. Notably, 1 disrupted the interaction between NDRG1 and c-Myc in CRC cells. In a mouse model with HCT116-implanted xenografts, the antitumor activity of 1 was confirmed by NDRG1 modulation. Overall, these findings show that 1 is a potential candidate for CRC treatment through regulation of NDGR1-mediated functionality.
Collapse
Affiliation(s)
- Kay Zin Kyaw
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Woong Sub Byun
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Yern-Hyerk Shin
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Thanh-Hau Huynh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji Yun Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun Seo Bae
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyen Joo Park
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Dong-Chan Oh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Sang Kook Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
8
|
Nozaki I, Ishikawa N, Miyanari Y, Ogawa K, Tagawa A, Yoshida S, Munekane M, Mishiro K, Toriba A, Nakayama M, Fuchigami T. Borealin-Derived Peptides as Survivin-Targeting Cancer Imaging and Therapeutic Agents. Bioconjug Chem 2022; 33:2149-2160. [DOI: 10.1021/acs.bioconjchem.2c00398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Iori Nozaki
- Laboratory of Clinical Analytical Sciences, Graduate School of Medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa920-1192, Japan
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki852-8521, Japan
| | - Natsumi Ishikawa
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki852-8521, Japan
| | - Yusuke Miyanari
- Institute of Nano Life Science, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa920-1192, Japan
| | - Kazuma Ogawa
- Laboratory of Clinical Analytical Sciences, Graduate School of Medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa920-1192, Japan
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa920-1192, Japan
| | - Ayako Tagawa
- Institute of Nano Life Science, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa920-1192, Japan
| | - Sakura Yoshida
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki852-8521, Japan
| | - Masayuki Munekane
- Laboratory of Clinical Analytical Sciences, Graduate School of Medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa920-1192, Japan
| | - Kenji Mishiro
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa920-1192, Japan
| | - Akira Toriba
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki852-8521, Japan
| | - Morio Nakayama
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki852-8521, Japan
| | - Takeshi Fuchigami
- Laboratory of Clinical Analytical Sciences, Graduate School of Medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa920-1192, Japan
| |
Collapse
|
9
|
Lisboa RV, de Oliveira FR, Quaresma TO, de Almeida RM, Ribeiro Oliveira RD, Junior PL. The Behaviour of Serum Survivin in Patients With Lupus Nephritis. Biomark Insights 2022; 17:11772719221131470. [PMID: 36311208 PMCID: PMC9597205 DOI: 10.1177/11772719221131470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 09/21/2022] [Indexed: 11/07/2022] Open
Abstract
Background: Systemic lupus erythematosus (SLE) is a chronic, multi phenotypic, autoimmune
inflammatory disease and renal involvement significantly worsens its
prognosis. Apoptosis dysregulation plays a key pathogenic role. Survivin, a
protein from the apoptosis inhibitors family, has been considered a
promising strategy in cancer therapy and evaluated as one of the regulatory
pathways in the scenario of immune-mediated disorders. Objective: This study aims to explore survivin behaviour in SLE patients with lupus
nephritis (LN), assessing its potential as a therapeutic and prognostic
biomarker. Methods: 297 SLE patients were classified based on the American College of
Rheumatology (ACR) 1997 criteria, from 2000 to 2015. In a cross-sectional
study, the serum level of survivin was measured by an ELISA test and
compared between 200 SLE individuals and healthy controls. In a longitudinal
cohort, 97 patients with active LN had the concentration of survinin
measured, before and after treatment with cyclophosphamide pulse
therapy. Results: The serum concentration of survivin was significantly lower in the SLE group
than in healthy controls, regardless of concomitant NL or disease activity.
The longitudinal evaluation revealed a significant reduction in survivin
serum level after treatment. However, survivin rates were not able to
discriminate groups that achieved remission from those that maintained
nephritis activity. Conclusion: Our study suggests that survivin levels in SLE patients are lower than in the
general population. Even so, its use as a biomarker in SLE seems limited,
not reflecting disease activity or response to LN treatment, as in other
contexts.
Collapse
Affiliation(s)
- Renata Valente Lisboa
- Ribeirão Preto Medical School, Ribeirão
Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil,Center of Research in Inflammatory
Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão
Preto, São Paulo, Brazil,Renata Valente Lisboa, Division of
Rheumatology, Ribeirão Preto Medical School, University of São Paulo, Av.
Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil.
| | - Fabiola Reis de Oliveira
- Ribeirão Preto Medical School, Ribeirão
Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil,Center of Research in Inflammatory
Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão
Preto, São Paulo, Brazil
| | - Thaise Oliveira Quaresma
- Ribeirão Preto Medical School, Ribeirão
Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil,Center of Research in Inflammatory
Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão
Preto, São Paulo, Brazil
| | | | - Rene Donizeti Ribeiro Oliveira
- Ribeirão Preto Medical School, Ribeirão
Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil,Center of Research in Inflammatory
Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão
Preto, São Paulo, Brazil
| | - Paulo Louzada Junior
- Ribeirão Preto Medical School, Ribeirão
Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil,Center of Research in Inflammatory
Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão
Preto, São Paulo, Brazil
| |
Collapse
|
10
|
Pandey SK, Shteinfer-Kuzmine A, Chalifa-Caspi V, Shoshan-Barmatz V. Non-apoptotic activity of the mitochondrial protein SMAC/Diablo in lung cancer: Novel target to disrupt survival, inflammation, and immunosuppression. Front Oncol 2022; 12:992260. [PMID: 36185255 PMCID: PMC9515501 DOI: 10.3389/fonc.2022.992260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial SMAC/Diablo induces apoptosis by binding the inhibitor of apoptosis proteins (IAPs), thereby activating caspases and, subsequently, apoptosis. Previously, we found that despite its pro-apoptotic activity, SMAC/Diablo is overexpressed in cancer, and demonstrated that in cancer it possesses new essential and non-apoptotic functions that are associated with regulating phospholipid synthesis including modulating mitochondrial phosphatidylserine decarboxylase activity. Here, we demonstrate additional functions for SMAC/Diablo associated with inflammation and immunity. CRISPR/Cas9 SMAC/Diablo-depleted A549 lung cancer cells displayed inhibited cell proliferation and migration. Proteomics analysis of these cells revealed altered expression of proteins associated with lipids synthesis and signaling, vesicular transport and trafficking, metabolism, epigenetics, the extracellular matrix, cell signaling, and neutrophil-mediated immunity. SMAC-KO A549 cell-showed inhibited tumor growth and proliferation and activated apoptosis. The small SMAC-depleted “tumor” showed a morphology of alveoli-like structures, reversed epithelial-mesenchymal transition, and altered tumor microenvironment. The SMAC-lacking tumor showed reduced expression of inflammation-related proteins such as NF-kB and TNF-α, and of the PD-L1, associated with immune system suppression. These results suggest that SMAC is involved in multiple processes that are essential for tumor growth and progression. Thus, targeting SMAC’s non-canonical function is a potential strategy to treat cancer.
Collapse
Affiliation(s)
- Swaroop Kumar Pandey
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anna Shteinfer-Kuzmine
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Vered Chalifa-Caspi
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Varda Shoshan-Barmatz
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- *Correspondence: Varda Shoshan-Barmatz,
| |
Collapse
|
11
|
Martínez-Sifuentes MA, Bassol-Mayagoitia S, Nava-Hernández MP, Ruiz-Flores P, Ramos-Treviño J, Haro-Santa Cruz J, Hernández-Ibarra JA. Survivin in Breast Cancer: A Review. Genet Test Mol Biomarkers 2022; 26:411-421. [PMID: 36166738 DOI: 10.1089/gtmb.2021.0286] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Breast cancer is the most frequently diagnosed cancer in women and ranks second among causes for cancer-related death in women. Gene technology has led to the recognition that breast cancer is a heterogeneous disease composed of different biological subtypes, and genetic profiling enables the response to chemotherapy to be predicted. This fact emphasizes the importance of selecting sensitive diagnostic and prognostic markers in the early disease stage and more efficient targeted treatments for this disease. One such prognostic marker appears to be survivin. Many studies have shown that survivin is strongly expressed in different types of cancers. Its overexpression has been demonstrated in breast cancer, and high activity of the survivin gene has been associated with a poor prognosis and worse survival rates.
Collapse
Affiliation(s)
- Manuel Antonio Martínez-Sifuentes
- Department of Reproductive Biology and Biomedical Research Center, School of Medicine, Autonomous University of Coahuila, Torreón, Mexico
| | - Susana Bassol-Mayagoitia
- Department of Reproductive Biology and Biomedical Research Center, School of Medicine, Autonomous University of Coahuila, Torreón, Mexico
| | - Martha P Nava-Hernández
- Department of Reproductive Biology and Biomedical Research Center, School of Medicine, Autonomous University of Coahuila, Torreón, Mexico
| | - Pablo Ruiz-Flores
- Department of Genetics and Molecular Medicine, Biomedical Research Center, School of Medicine, Autonomous University of Coahuila, Torreón, Mexico
| | - Juan Ramos-Treviño
- Department of Reproductive Biology and Biomedical Research Center, School of Medicine, Autonomous University of Coahuila, Torreón, Mexico
| | - Jorge Haro-Santa Cruz
- Department of Genetics and Molecular Medicine, Biomedical Research Center, School of Medicine, Autonomous University of Coahuila, Torreón, Mexico
| | - José Anselmo Hernández-Ibarra
- Department of Reproductive Biology and Biomedical Research Center, School of Medicine, Autonomous University of Coahuila, Torreón, Mexico
| |
Collapse
|
12
|
de Oliveira LCB, Ribeiro DL, do Nascimento JR, da Rocha CQ, de Syllos Cólus IM, Serpeloni JM. Anticancer activities of Brachydin C in human prostate tumor cells (DU145) grown in 2D and 3D models: stimulation of cell death and downregulation of metalloproteinases in spheroids. Chem Biol Drug Des 2022; 100:747-762. [PMID: 35775856 DOI: 10.1111/cbdd.14112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/24/2022] [Accepted: 06/26/2022] [Indexed: 11/27/2022]
Abstract
Brachydin C (BrC) has demonstrated in vitro cytotoxic and antiproliferative effects in prostate cancer cells. In the present study, we compare the anticancer effects of BrC in DU145 cells grown in common bidimensional cultures (2D) and multicellular tumor spheroids (MCTS), often denominated 3D in vitro models, that can better mimic the microenvironment of tissues. BrC IC50 values obtained in the resazurin assay after 24 h of treatment were 47.31 μM (2D) and 229.8 μM (3D) and these cytotoxic effects were time dependent only in 3D. BrC (5 to 60 μM) interfered with the growth of MCTS and reduced cell viability after 11 days of treatment, a result that is not attributable to oxidative stress evaluated using the CM-H2 DCFDA probe. BrC (6.0 μM) impaired horizontal (wound healing) and vertical cell migration and invasion (transwell assay) in 2D and BrC (5.0 to 60 μM) in 3D (ECM Gel®). BrC modulated the expression of genes BIRC5, TNF-α, CASP3, NKX3.1, MMP9, MMP11, CDH1, and ITGAM and downregulated proteins CASP7, BAX, and TNF-α in western blotting analysis. In conclusion, BrC stimulated cell death and decreased epithelial-mesenchymal transition. Furthermore, DU145 MCTS displayed higher resistance to BrC- induced cell death than 2D cultures, a difference that should be considered in future approaches in prostatic cancer studies.
Collapse
Affiliation(s)
| | - Diego Luis Ribeiro
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | | | - Claudia Quintino da Rocha
- Department of Chemistry, Center for Exact Sciences and Technology, Federal University of Maranhão, São Luís, Brazil
| | - Ilce Mara de Syllos Cólus
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina, Brazil
| | - Juliana Mara Serpeloni
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina, Brazil
| |
Collapse
|
13
|
Resveratrol as a modulatory of apoptosis and autophagy in cancer therapy. Clin Transl Oncol 2022; 24:1219-1230. [PMID: 35038152 DOI: 10.1007/s12094-021-02770-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/24/2021] [Indexed: 12/24/2022]
Abstract
Cancer is one of the leading causes of death, with a heavy socio-economical burden for countries. Despite the great advances that have been made in the treatment of cancer, chemotherapy is still the most common method of treatment. However, many side effects, including hepatotoxicity, renal toxicity, and cardiotoxicity, limit the efficacy of conventional chemotherapy. Over recent years, natural products have attracted attention as therapeutic agents against various diseases, such as cancer. Resveratrol (RES), a natural polyphenol occurring in grapes, nuts, wine, and berries, exhibited potential for preventing and treating various cancer types. RES also ameliorates chemotherapy-induced detrimental effects. Furthermore, RES could modulate apoptosis and autophagy as the main forms of cancer cell deaths by targeting various signaling pathways and up/downregulation of apoptotic and autophagic genes. This review will summarize the anti-cancer effects of RES and focus on the fundamental mechanisms and targets for modulating apoptosis and autophagy by RES.
Collapse
|
14
|
Xu S, Wang X, Fei C. A Highly Effective System for Predicting MHC-II Epitopes With Immunogenicity. Front Oncol 2022; 12:888556. [PMID: 35785204 PMCID: PMC9246415 DOI: 10.3389/fonc.2022.888556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/27/2022] [Indexed: 12/30/2022] Open
Abstract
In the past decade, the substantial achievements of therapeutic cancer vaccines have shed a new light on cancer immunotherapy. The major challenge for designing potent therapeutic cancer vaccines is to identify neoantigens capable of inducing sufficient immune responses, especially involving major histocompatibility complex (MHC)-II epitopes. However, most previous studies on T-cell epitopes were focused on either ligand binding or antigen presentation by MHC rather than the immunogenicity of T-cell epitopes. In order to better facilitate a therapeutic vaccine design, in this study, we propose a revolutionary new tool: a convolutional neural network model named FIONA (Flexible Immunogenicity Optimization Neural-network Architecture) trained on IEDB datasets. FIONA could accurately predict the epitopes presented by the given specific MHC-II subtypes, as well as their immunogenicity. By leveraging the human leukocyte antigen allele hierarchical encoding model together with peptide dense embedding fusion encoding, FIONA (with AUC = 0.94) outperforms several other tools in predicting epitopes presented by MHC-II subtypes in head-to-head comparison; moreover, FIONA has unprecedentedly incorporated the capacity to predict the immunogenicity of epitopes with MHC-II subtype specificity. Therefore, we developed a reliable pipeline to effectively predict CD4+ T-cell immune responses against cancer and infectious diseases.
Collapse
Affiliation(s)
| | | | - Caiyi Fei
- Department of AI and Bioinformatics, Nanjing Chengshi BioTech (TheraRNA) Co., Ltd., Nanjing, China
| |
Collapse
|
15
|
Shomali N, Suliman Maashi M, Baradaran B, Daei Sorkhabi A, Sarkesh A, Mohammadi H, Hemmatzadeh M, Marofi F, Sandoghchian Shotorbani S, Jarahian M. Dysregulation of Survivin-Targeting microRNAs in Autoimmune Diseases: New Perspectives for Novel Therapies. Front Immunol 2022; 13:839945. [PMID: 35309327 PMCID: PMC8927965 DOI: 10.3389/fimmu.2022.839945] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/14/2022] [Indexed: 12/15/2022] Open
Abstract
It has been well established that the etiopathogenesis of diverse autoimmune diseases is rooted in the autoreactive immune cells' excessively proliferative state and impaired apoptotic machinery. Survivin is an anti-apoptotic and mitotic factor that has sparked a considerable research interest in this field. Survivin overexpression has been shown to contribute significantly to the development of autoimmune diseases via autoreactive immune cell overproliferation and apoptotic dysregulation. Several microRNAs (miRNAs/miRs) have been discovered to be involved in survivin regulation, rendering the survivin-miRNA axis a perspective target for autoimmune disease therapy. In this review, we discuss the role of survivin as an immune regulator and a highly implicated protein in the pathogenesis of autoimmune diseases, the significance of survivin-targeting miRNAs in autoimmunity, and the feasibility of targeting the survivin-miRNA axis as a promising therapeutic option for autoimmune diseases.
Collapse
Affiliation(s)
- Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marwah Suliman Maashi
- Medical Laboratory Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Daei Sorkhabi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aila Sarkesh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Maryam Hemmatzadeh
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siamak Sandoghchian Shotorbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy Unit (G401), Heidelberg, Germany
| |
Collapse
|
16
|
Tasdemir-Yilmaz OE, Druckenbrod NR, Olukoya OO, Dong W, Yung AR, Bastille I, Pazyra-Murphy MF, Sitko AA, Hale EB, Vigneau S, Gimelbrant AA, Kharchenko PV, Goodrich LV, Segal RA. Diversity of developing peripheral glia revealed by single-cell RNA sequencing. Dev Cell 2021; 56:2516-2535.e8. [PMID: 34469751 DOI: 10.1016/j.devcel.2021.08.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 05/31/2021] [Accepted: 08/06/2021] [Indexed: 12/22/2022]
Abstract
The peripheral nervous system responds to a wide variety of sensory stimuli, a process that requires great neuronal diversity. These diverse neurons are closely associated with glial cells originating from the neural crest. However, the molecular nature and diversity among peripheral glia are not understood. Here, we used single-cell RNA sequencing to profile developing and mature glia from somatosensory dorsal root ganglia and auditory spiral ganglia. We found that glial precursors (GPs) in these two systems differ in their transcriptional profiles. Despite their unique features, somatosensory and auditory GPs undergo convergent differentiation to generate molecularly uniform myelinating and non-myelinating Schwann cells. By contrast, somatosensory and auditory satellite glial cells retain system-specific features. Lastly, we identified a glial signature gene set, providing new insights into commonalities among glia across the nervous system. This survey of gene expression in peripheral glia constitutes a resource for understanding functions of glia across different sensory modalities.
Collapse
Affiliation(s)
- Ozge E Tasdemir-Yilmaz
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Noah R Druckenbrod
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Weixiu Dong
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Andrea R Yung
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Isle Bastille
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Maria F Pazyra-Murphy
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Austen A Sitko
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Evan B Hale
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Sébastien Vigneau
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Peter V Kharchenko
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Rosalind A Segal
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
17
|
Zhou WT, Jin WL. B7-H3/CD276: An Emerging Cancer Immunotherapy. Front Immunol 2021; 12:701006. [PMID: 34349762 PMCID: PMC8326801 DOI: 10.3389/fimmu.2021.701006] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/05/2021] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy aiming at suppressing tumor development by relying on modifying or strengthening the immune system prevails among cancer treatments and points out a new direction for cancer therapy. B7 homolog 3 protein (B7-H3, also known as CD276), a newly identified immunoregulatory protein member of the B7 family, is an attractive and promising target for cancer immunotherapy because it is overexpressed in tumor tissues while showing limited expression in normal tissues and participating in tumor microenvironment (TME) shaping and development. Thus far, numerous B7-H3-based immunotherapy strategies have demonstrated potent antitumor activity and acceptable safety profiles in preclinical models. Herein, we present the expression and biological function of B7-H3 in distinct cancer and normal cells, as well as B7-H3-mediated signal pathways in cancer cells and B7-H3-based tumor immunotherapy strategies. This review provides a comprehensive overview that encompasses B7-H3’s role in TME to its potential as a target in cancer immunotherapy.
Collapse
Affiliation(s)
- Wu-Tong Zhou
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, China.,Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| |
Collapse
|
18
|
Núñez-Iglesias MJ, Novio S, García C, Pérez-Muñuzuri ME, Martínez MC, Santiago JL, Boso S, Gago P, Freire-Garabal M. Co-Adjuvant Therapy Efficacy of Catechin and Procyanidin B2 with Docetaxel on Hormone-Related Cancers In Vitro. Int J Mol Sci 2021; 22:7178. [PMID: 34281228 PMCID: PMC8268784 DOI: 10.3390/ijms22137178] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 01/16/2023] Open
Abstract
Prostate (PC) and breast cancer (BC) are heterogeneous hormonal cancers. Treatment resistance and adverse effects are the main limitations of conventional chemotherapy treatment. The use of sensitizing agents could improve the effectiveness of chemotherapeutic drugs as well as obviate these limitations. This study analyzes the effect of single catechin (CAT), procyanidin B2 (ProB2) treatment as well as the co-adjuvant treatment of each of these compounds with docetaxel (DOCE). We used PC- and BC-derived cell lines (PC3, DU-145, T47D, MCF-7 and MDA-MB-231). The short and long-term pro-apoptotic, anti-proliferative and anti-migratory effects were analyzed. RT-qPCR was used to discover molecular bases of the therapeutic efficacy of these compounds. ProB2 treatment induced a two- to five-fold increase in anti-proliferative and pro-apoptotic effects compared to single DOCE treatment, and also had a more sensitizing effect than DOCE on DU145 cells. Regarding BC cells, ProB2- and CAT-mediated sensitization to DOCE anti-proliferative and pro-apoptotic effects was cell-independent and cell-dependent, respectively. Combined treatment led to high-efficacy effects on MCF-7 cells, which were associated to the up-regulation of CDKN1A, BAX, caspase 9 and E-cadherin mRNA under combined treatment compared to single DOCE treatment. CAT and ProB2 can enhance the efficacy of DOCE therapy on PC and BC cells by the sensitizing mechanism.
Collapse
Affiliation(s)
- Mª Jesús Núñez-Iglesias
- SNL Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, Santiago de Compostela, 15782 A Coruña, Spain; (M.J.N.-I.); (C.G.); (M.E.P.-M.); (M.F.-G.)
| | - Silvia Novio
- SNL Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, Santiago de Compostela, 15782 A Coruña, Spain; (M.J.N.-I.); (C.G.); (M.E.P.-M.); (M.F.-G.)
| | - Carlota García
- SNL Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, Santiago de Compostela, 15782 A Coruña, Spain; (M.J.N.-I.); (C.G.); (M.E.P.-M.); (M.F.-G.)
| | - Mª Elena Pérez-Muñuzuri
- SNL Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, Santiago de Compostela, 15782 A Coruña, Spain; (M.J.N.-I.); (C.G.); (M.E.P.-M.); (M.F.-G.)
| | - María-Carmen Martínez
- Group of Viticulture, Olive and Rose (VIOR), Misión Biológica de Galicia, Consejo Superior de Investigaciones Científicas (CSIC), Carballeira 8, 36143 Salcedo, Spain; (M.-C.M.); (J.-L.S.); (S.B.); (P.G.)
| | - José-Luis Santiago
- Group of Viticulture, Olive and Rose (VIOR), Misión Biológica de Galicia, Consejo Superior de Investigaciones Científicas (CSIC), Carballeira 8, 36143 Salcedo, Spain; (M.-C.M.); (J.-L.S.); (S.B.); (P.G.)
| | - Susana Boso
- Group of Viticulture, Olive and Rose (VIOR), Misión Biológica de Galicia, Consejo Superior de Investigaciones Científicas (CSIC), Carballeira 8, 36143 Salcedo, Spain; (M.-C.M.); (J.-L.S.); (S.B.); (P.G.)
| | - Pilar Gago
- Group of Viticulture, Olive and Rose (VIOR), Misión Biológica de Galicia, Consejo Superior de Investigaciones Científicas (CSIC), Carballeira 8, 36143 Salcedo, Spain; (M.-C.M.); (J.-L.S.); (S.B.); (P.G.)
| | - Manuel Freire-Garabal
- SNL Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, Santiago de Compostela, 15782 A Coruña, Spain; (M.J.N.-I.); (C.G.); (M.E.P.-M.); (M.F.-G.)
| |
Collapse
|
19
|
Jasek-Gajda E, Jurkowska H, JasiŃska M, Litwin JA, Lis GJ. Combination of ERK2 and STAT3 Inhibitors Promotes Anticancer Effects on Acute Lymphoblastic Leukemia Cells. Cancer Genomics Proteomics 2021; 17:517-527. [PMID: 32859630 DOI: 10.21873/cgp.20208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/01/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND/AIM Deregulated activation of signaling through the RAS/RAF/mitogen-activated protein kinase/extracellular signal-regulated kinase (RAS/RAF/MEK/ERK) and signal transducer and activator of transcription (STAT) pathways is involved in numerous hematological malignancies, making it an attractive therapeutic target. This study aimed to assess the effect of the combination of ERK2 inhibitor VX-11e and STAT3 inhibitor STA-21 on acute lymphoblastic leukemia cell lines REH and MOLT-4. MATERIALS AND METHODS REH and MOLT-4 cell lines were cultured with each drug alone and in combination. Cell viability, ERK activity, cell cycle distribution, apoptosis and oxidative stress induction were assessed by flow cytometry. Protein levels of STAT3, phospho-STAT3, protein tyrosine phosphatase 4A3 (PTP4A3), survivin, p53 and p21 were determined by western blotting. RESULTS VX-11e in combination with STA-21 significantly inhibited cell viability, induced G0/G1 cell-cycle arrest, enhanced production of reactive oxygen species, and induced apoptosis. These effects were associated with an increased level of p21 protein in REH cells and with reduced levels of phopho-STAT3, survivin and PTP4A3 proteins in MOLT-4 cells. CONCLUSION Our findings provide a rationale for combined inhibition of RAS/RAF/MEK/ERK and STAT3 pathways in order to enhance anticancer effects against acute lymphoblastic leukemia cells.
Collapse
Affiliation(s)
- Ewa Jasek-Gajda
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Halina Jurkowska
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - MaŁgorzata JasiŃska
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Jan A Litwin
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Grzegorz J Lis
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
20
|
Hill J, Zawia NH. Fenamates as Potential Therapeutics for Neurodegenerative Disorders. Cells 2021; 10:702. [PMID: 33809987 PMCID: PMC8004804 DOI: 10.3390/cells10030702] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 11/16/2022] Open
Abstract
Neurodegenerative disorders are desperately lacking treatment options. It is imperative that drug repurposing be considered in the fight against neurodegenerative diseases. Fenamates have been studied for efficacy in treating several neurodegenerative diseases. The purpose of this review is to comprehensively present the past and current research on fenamates in the context of neurodegenerative diseases with a special emphasis on tolfenamic acid and Alzheimer's disease. Furthermore, this review discusses the major molecular pathways modulated by fenamates.
Collapse
Affiliation(s)
- Jaunetta Hill
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881, USA;
| | - Nasser H. Zawia
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881, USA;
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI 02881, USA
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
21
|
Integrative Transcriptomic Network Analysis of Butyrate Treated Colorectal Cancer Cells. Cancers (Basel) 2021; 13:cancers13040636. [PMID: 33562636 PMCID: PMC7914650 DOI: 10.3390/cancers13040636] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 01/14/2023] Open
Abstract
Diet-derived histone deacetylase inhibitor (HDACi), butyrate, alters global acetylation and consequently global gene expression in colorectal cancer (CRC) cells to exert its anticancer effects. Aberrant microRNA (miRNA) expression contributes to CRC development and progression. Butyrate-mediated modulation of microRNA (miRNA) expression remains under-investigated. This study employed a systems biology approach to gain a comprehensive understanding of the complex miRNA-mRNA interactions contributing to the butyrate response in CRC cells. Next-generation sequencing, gene ontology (GO) and pathway enrichment analyses were utilized to reveal the extent of butyrate-mediated gene regulation in CRC cells. Changes in cell proliferation, apoptosis, the cell cycle and gene expression induced by miRNAs and target gene knockdown in CRC cells were assessed. Butyrate induced differential expression of 113 miRNAs and 2447 protein-coding genes in HCT116 cells. Butyrate also altered transcript splicing of 1591 protein-coding genes. GO, and pathway enrichment analyses revealed the cell cycle to be a central target of the butyrate response. Two butyrate-induced miRNAs, miR-139 and miR-542, acted cooperatively with butyrate to induce apoptosis and reduce CRC cell proliferation by regulating target genes, including cell cycle-related EIF4G2 and BIRC5. EIF4G2 RNA interference mimicked the miR-139-mediated reduction in cell proliferation. The cell cycle is a critical pathway involved in the butyrate response of CRC cells. These findings reveal novel roles for miRNAs in the cell cycle-related, anticancer effects of butyrate in CRC cells.
Collapse
|
22
|
Meier T, Timm M, Montani M, Wilkens L. Gene networks and transcriptional regulators associated with liver cancer development and progression. BMC Med Genomics 2021; 14:41. [PMID: 33541355 PMCID: PMC7863452 DOI: 10.1186/s12920-021-00883-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 01/24/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Treatment options for hepatocellular carcinoma (HCC) are limited, and overall survival is poor. Despite the high frequency of this malignoma, its basic disease mechanisms are poorly understood. Therefore, the aim of this study was to use different methodological approaches and combine the results to improve our knowledge on the development and progression of HCC. METHODS Twenty-three HCC samples were characterized by histological, morphometric and cytogenetic analyses, as well as comparative genomic hybridization (aCGH) and genome-wide gene expression followed by a bioinformatic search for potential transcriptional regulators and master regulatory molecules of gene networks. RESULTS Histological evaluation revealed low, intermediate and high-grade HCCs, and gene expression analysis split them into two main sets: GE1-HCC and GE2-HCC, with a low and high proliferation gene expression signature, respectively. Array-based comparative genomic hybridization demonstrated a high level of chromosomal instability, with recurrent chromosomal gains of 1q, 6p, 7q, 8q, 11q, 17q, 19p/q and 20q in both HCC groups and losses of 1p, 4q, 6q, 13q and 18q characteristic for GE2-HCC. Gene expression and bioinformatics analyses revealed that different genes and gene regulatory networks underlie the distinct biological features observed in GE1-HCC and GE2-HCC. Besides previously reported dysregulated genes, the current study identified new candidate genes with a putative role in liver cancer, e.g. C1orf35, PAFAH1B3, ZNF219 and others. CONCLUSION Analysis of our findings, in accordance with the available published data, argues in favour of the notion that the activated E2F1 signalling pathway, which can be responsible for both inappropriate cell proliferation and initial chromosomal instability, plays a pivotal role in HCC development and progression. A dedifferentiation switch that manifests in exaggerated gene expression changes might be due to turning on transcriptional co-regulators with broad impact on gene expression, e.g. POU2F1 (OCT1) and NFY, as a response to accumulating cell stress during malignant development. Our findings point towards the necessity of different approaches for the treatment of HCC forms with low and high proliferation signatures and provide new candidates for developing appropriate HCC therapies.
Collapse
Affiliation(s)
- Tatiana Meier
- Institute of Pathology, Nordstadtkrankenhaus, Hanover, Germany.
| | - Max Timm
- Institute of Pathology, Nordstadtkrankenhaus, Hanover, Germany
- Clinic for Laryngology, Rhinology and Otology, Medical School Hanover, Hanover, Germany
| | - Matteo Montani
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Ludwig Wilkens
- Institute of Pathology, Nordstadtkrankenhaus, Hanover, Germany
- Institute of Human Genetics, Medical School Hanover, Hanover, Germany
| |
Collapse
|
23
|
Ambrose AJ, Pham NT, Sivinski J, Guimarães L, Mollasalehi N, Jimenez P, Abad MA, Jeyaprakash AA, Shave S, Costa-Lotufo LV, La Clair JJ, Auer M, Chapman E. A two-step resin based approach to reveal survivin-selective fluorescent probes. RSC Chem Biol 2021; 2:181-186. [PMID: 34458780 PMCID: PMC8342005 DOI: 10.1039/d0cb00122h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 11/07/2020] [Indexed: 01/24/2023] Open
Abstract
The identification of modulators for proteins without assayable biochemical activity remains a challenge in chemical biology. The presented approach adapts a high-throughput fluorescence binding assay and functional chromatography, two protein-resin technologies, enabling the discovery and isolation of fluorescent natural product probes that target proteins independently of biochemical function. The resulting probes also suggest targetable pockets for lead discovery. Using human survivin as a model, we demonstrate this method with the discovery of members of the prodiginine family as fluorescent probes to the cancer target survivin.
Collapse
Affiliation(s)
- Andrew J Ambrose
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona Tucson AZ 85721 USA
| | - Nhan T Pham
- School of Biological Sciences and Edinburgh Medical School, Biomedical Sciences, University of Edinburgh The King's Buildings CH Waddington Building 3.07 Max Born Crescent Edinburgh EH9 3BF UK
| | - Jared Sivinski
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona Tucson AZ 85721 USA
| | - Larissa Guimarães
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona Tucson AZ 85721 USA
- Departamento de Farmacologia, Universidade de São Paulo São Paulo SP 05508-900 Brazil
| | - Niloufar Mollasalehi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona Tucson AZ 85721 USA
| | - Paula Jimenez
- Instituto do Mar, Universidade Federal de São Paulo Santos SP 11.070-100 Brazil
| | - Maria A Abad
- Wellcome Centre for Cell Biology, University of Edinburgh Edinburgh EH9 3BF UK
| | | | - Steven Shave
- School of Biological Sciences and Edinburgh Medical School, Biomedical Sciences, University of Edinburgh The King's Buildings CH Waddington Building 3.07 Max Born Crescent Edinburgh EH9 3BF UK
| | | | - James J La Clair
- Xenobe Research Institute P. O. Box 3052 San Diego CA 92163-1052 USA
| | - Manfred Auer
- School of Biological Sciences and Edinburgh Medical School, Biomedical Sciences, University of Edinburgh The King's Buildings CH Waddington Building 3.07 Max Born Crescent Edinburgh EH9 3BF UK
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona Tucson AZ 85721 USA
| |
Collapse
|
24
|
Survivin Regulates Bad Gene Expression by Binding to Its Promoter and Modulates Cell Cycle and Apoptosis in Esophageal Carcinoma Cell. JOURNAL OF ONCOLOGY 2021; 2021:1384289. [PMID: 33488710 PMCID: PMC7803392 DOI: 10.1155/2021/1384289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 11/24/2022]
Abstract
Esophageal cancer (EC) is the eighth most prevalent cancer and the sixth leading cause of cancer-related mortality worldwide. As an antiapoptotic and a proapoptotic protein, respectively, survivin and Bad play an important role in carcinogenesis of the most human cancers including EC. However, the regulatory relationships between them remain unclear. We sought to investigate the effects of survivin knockdown and overexpression on the expression of Bad gene, cell cycle progression, and apoptosis of esophageal carcinoma cell. The mRNA expression levels of survivin and Bad were determined in EC tissue samples. The knockdown and overexpression experiments were performed in ECA109 and KYSE450 cells via transfection with survivin overexpression and shRNA plasmids. A Bad overexpression experiment was conducted to confirm the biological effect on knockdown of survivin via modulating Bad expression. RT-qPCR and Western blot analysis were used to detect mRNA and protein expression, respectively. Cell cycle and apoptosis were analyzed by flow cytometry. The chromatin immunoprecipitation (ChIP) was conducted to determine the binding sites of survivin on the promoter of Bad gene. By analyzing the mRNA expression of survivin and Bad in 40 ESCC patient specimens, we found that the positive expression rate of survivin in tumor tissues (88%, 35/40) was remarkably high, compared with the distal nontumor tissues (48%, 19/40, p < 0.01). On the other hand, the positive expression rate of Bad in tumor tissues (70%, 28/40) was remarkably low, compared with the distal nontumor tissues (95%, 38/40, p < 0.01). Overexpression of survivin decreases Bad mRNA and protein expression and promotes transformation of cell cycle to S phase. Conversely, knockdown of survivin increases Bad mRNA and protein expression and induces cell cycle arrest and apoptosis. Bad overexpression inducing apoptosis of esophageal carcinoma cell shows the similar apoptotic effect with survivin knockdown. ChIP assays indicate that survivin directly binds to the Bad promoter region, diminishing the transcriptional activity of Bad. In conclusion, the result suggested that survivin regulates Bad gene expression by binding to its promoter and modulates cell cycle and apoptosis in esophageal carcinoma cell.
Collapse
|
25
|
Sari C, SÜmer C, Celep EyÜpoĞlu F. Caffeic acid phenethyl ester induces apoptosis in colorectal cancer cells via inhibition of survivin. ACTA ACUST UNITED AC 2020; 44:264-274. [PMID: 33110364 PMCID: PMC7585156 DOI: 10.3906/biy-2003-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/18/2020] [Indexed: 01/08/2023]
Abstract
Colorectal cancer is one of the most common types of cancer. Drug resistance and drug-induced damage of healthy tissues are major obstacles in cancer treatment. Therefore, to develop efficient anticancer therapy, it is necessary to find compounds that affect tumor cells, but do not exhibit toxicity to healthy cells. Caffeic acid phenethyl ester (CAPE) has been demonstrated to have anticancer properties in many types of cancer. In this study, the cytotoxic and apoptotic effects of CAPE on the RKO colorectal cancer cell line and CCD 841-CoN normal colorectal cell line was investigated. In addition, changes in the survivin expression were determined. According to the results, CAPE decreased cell viability in the RKO cell line in a dose-dependent manner. Likewise, CAPE induced apoptotic cell death in approximately 40% of the RKO cells. Furthermore, CAPE treatment increased the Serine 15 (Ser15) and Serine 46 (Ser46) phosphorylation of p53, while decreased the survivin expression. The results suggested that CAPE induced apoptosis by regulating p53 phosphorylation, leading to inhibition of the survivin expression. In accordance with the results, it is suggested that CAPE might be evaluated as an alternative drug in cancer therapy and further investigation is needed within this scope.
Collapse
Affiliation(s)
- Ceren Sari
- Department of Medical Biology, Institute of Health Sciences, Karadeniz Technical University, Trabzon Turkey
| | - Ceren SÜmer
- Department of Medical Biology, Institute of Health Sciences, Karadeniz Technical University, Trabzon Turkey
| | - Figen Celep EyÜpoĞlu
- Department of Medical Biology, Faculty of Medicine, Karadeniz Technical University, Trabzon Turkey
| |
Collapse
|
26
|
Combination of ERK2 inhibitor VX-11e and voreloxin synergistically enhances anti-proliferative and pro-apoptotic effects in leukemia cells. Apoptosis 2020; 24:849-861. [PMID: 31482470 PMCID: PMC6823322 DOI: 10.1007/s10495-019-01564-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
ERK1/2 inhibitors are new promising anticancer drugs. The aim of this study was to investigate the effect of the combination of ERK2 inhibitor VX-11e and voreloxin on MOLM-14, K562, REH and MOLT-4 leukemia cell lines. We found that VX-11e alone and in combination with voreloxin significantly decreased ERK activation in all cell lines tested. To evaluate the interactions of the drugs, cells were treated for 24 h with VX-11e or voreloxin alone and in combination at fixed ratios based on IC50 values. The combinatorial effects of both drugs were synergistic over a wide range of concentrations in MOLM-14, REH and MOLT-4 cell lines. In K562 cells, three effects were found to be additive, one antagonistic and only one synergistic. The results showed that incubation with both VX-11e and voreloxin inhibited the growth of leukemia cells, affected cell cycle and induced apoptosis. Furthermore, the molecular mechanism of these effects might be attributed to an increased expression of p21 and a decreased expression of survivin and NF-κB in all cell lines tested except from K562 cells. In conclusion, combination of VX-11e and voreloxin can exert a synergistic anticancer effect in leukemia cells.
Collapse
|
27
|
Targeting the MAPK/ERK and PI3K/AKT Signaling Pathways Affects NRF2, Trx and GSH Antioxidant Systems in Leukemia Cells. Antioxidants (Basel) 2020; 9:antiox9070633. [PMID: 32709140 PMCID: PMC7402140 DOI: 10.3390/antiox9070633] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 12/31/2022] Open
Abstract
The mitogen-activated protein kinase (MAPK)/extracellular signal kinase (ERK) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signal transduction pathways have been implicated in the pathogenesis of leukemia. The aim of this study was to investigate the effect of the combination of ERK1/2 inhibitor AZD0364 and PI3K inhibitor ZSTK474 on acute lymphoblastic leukemia (ALL) REH, MOLT-4, acute myeloid leukemia (AML) MOLM-14, and chronic myeloid leukemia (CML) K562 cell lines. To evaluate the interactions of the drugs, cells were treated for 48 h with AZD0364 or ZSTK474 alone and in combination at fixed ratios. The combinatorial effects of both inhibitors were synergistic over a wide range of concentrations in REH, MOLT-4, and MOLM-14 cell lines. However, in K562 cells, the effects were found to be antagonistic. Furthermore, AZD0364 and ZSTK474 significantly decreased both ERK1/2 and AKT activation in REH, MOLT-4, and MOLM-14 cells. The results showed that incubation with both AZD0364 and ZSTK474 inhibited cell viability, increased reactive oxygen species (ROS) production, and induced apoptosis in leukemia cells. We observed that combined treatment with AZD0364 and ZSTK474 affected nuclear factor-κB (NF-κB) and antioxidant protein levels: NF-E2-related factor 2 (NRF2), heme oxygenase-1 (HO-1), thioredoxin (Trx), thioredoxin reductase (TrxR), and the reduced glutathione/oxidized glutathione (GSH/GSSG) ratio. These effects were accompanied with decreased antiapoptotic survivin protein level. However, distinct cell line dependent effects were observed. In conclusion, the combination of AZD0364 and ZSTK474 can exert a synergistic anticancer effect in ALL and AML cells, which is associated with the induction of oxidative stress and the involvement of cellular antioxidant defense mechanisms.
Collapse
|
28
|
León D, Buchegger K, Silva R, Riquelme I, Viscarra T, Mora-Lagos B, Zanella L, Schafer F, Kurachi C, Roa JC, Ili C, Brebi P. Epigallocatechin Gallate Enhances MAL-PDT Cytotoxic Effect on PDT-Resistant Skin Cancer Squamous Cells. Int J Mol Sci 2020; 21:ijms21093327. [PMID: 32397263 PMCID: PMC7247423 DOI: 10.3390/ijms21093327] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 02/06/2023] Open
Abstract
Photodynamic therapy (PDT) has been used to treat certain types of non-melanoma skin cancer with promising results. However, some skin lesions have not fully responded to this treatment, suggesting a potential PDT-resistant phenotype. Therefore, novel therapeutic alternatives must be identified that improve PDT in resistant skin cancer. In this study, we analyzed the cell viability, intracellular protoporphyrin IX (PpIX) content and subcellular localization, proliferation profile, cell death, reactive oxygen species (ROS) detection and relative gene expression in PDT-resistant HSC-1 cells. PDT-resistant HSC-1 cells show a low quantity of protoporphyrin IX and low levels of ROS, and thus a low rate of death cell. Furthermore, the resistant phenotype showed a downregulation of HSPB1, SLC15A2, FECH, SOD2 and an upregulation of HMBS and BIRC5 genes. On the other hand, epigallocatechin gallate catechin enhanced the MAL-PDT effect, increasing levels of protoporphyrin IX and ROS, and killing 100% of resistant cells. The resistant MAL-PDT model of skin cancer squamous cells (HSC-1) is a reliable and useful tool to understand PDT cytotoxicity and cellular response. These resistant cells were successfully sensitized with epigallocatechin gallate catechin. The in vitro epigallocatechin gallate catechin effect as an enhancer of MAL-PDT in resistant cells is promising in the treatment of difficult skin cancer lesions.
Collapse
Affiliation(s)
- Daniela León
- Laboratory of Integrative Biology, Centro de Excelencia en Medicina Traslacional—Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (D.L.); (K.B.); (T.V.); (B.M.-L.); (L.Z.)
| | - Kurt Buchegger
- Laboratory of Integrative Biology, Centro de Excelencia en Medicina Traslacional—Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (D.L.); (K.B.); (T.V.); (B.M.-L.); (L.Z.)
- Department of Basic Sciences, School of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
| | - Ramón Silva
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud. Universidad Autónoma de Chile, Temuco 4810101, Chile; (R.S.); (I.R.)
| | - Ismael Riquelme
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud. Universidad Autónoma de Chile, Temuco 4810101, Chile; (R.S.); (I.R.)
| | - Tamara Viscarra
- Laboratory of Integrative Biology, Centro de Excelencia en Medicina Traslacional—Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (D.L.); (K.B.); (T.V.); (B.M.-L.); (L.Z.)
| | - Bárbara Mora-Lagos
- Laboratory of Integrative Biology, Centro de Excelencia en Medicina Traslacional—Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (D.L.); (K.B.); (T.V.); (B.M.-L.); (L.Z.)
| | - Louise Zanella
- Laboratory of Integrative Biology, Centro de Excelencia en Medicina Traslacional—Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (D.L.); (K.B.); (T.V.); (B.M.-L.); (L.Z.)
| | - Fabiola Schafer
- Department of Medical Specialties, School of Medicine, Universidad de La Frontera, Temuco 4811230, Chile;
| | - Cristina Kurachi
- São Carlos Institute of Physics, University of São Paulo (USP), P.O. Box 369, São Carlos 13560-970, São Paulo, Brazil;
| | - Juan Carlos Roa
- Department of Pathology, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile;
| | - Carmen Ili
- Laboratory of Integrative Biology, Centro de Excelencia en Medicina Traslacional—Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (D.L.); (K.B.); (T.V.); (B.M.-L.); (L.Z.)
- Correspondence: (C.I.); (P.B.); Tel.: +56-45-2-596693 (C.I.); +56-45-2-596583 (P.B.)
| | - Priscilla Brebi
- Laboratory of Integrative Biology, Centro de Excelencia en Medicina Traslacional—Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (D.L.); (K.B.); (T.V.); (B.M.-L.); (L.Z.)
- Correspondence: (C.I.); (P.B.); Tel.: +56-45-2-596693 (C.I.); +56-45-2-596583 (P.B.)
| |
Collapse
|
29
|
Jordaens S, Cooksey L, Freire Boullosa L, Van Tendeloo V, Smits E, Mills KI, Orchard KH, Guinn BA. New targets for therapy: antigen identification in adults with B-cell acute lymphoblastic leukaemia. Cancer Immunol Immunother 2020; 69:867-877. [PMID: 31970440 PMCID: PMC7183504 DOI: 10.1007/s00262-020-02484-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 01/04/2020] [Indexed: 12/11/2022]
Abstract
Acute lymphoblastic leukaemia (ALL) in adults is a rare and difficult-to-treat cancer that is characterised by excess lymphoblasts in the bone marrow. Although many patients achieve remission with chemotherapy, relapse rates are high and the associated impact on survival devastating. Most patients receive chemotherapy and for those whose overall fitness supports it, the most effective treatment to date is allogeneic stem cell transplant that can improve overall survival rates in part due to a 'graft-versus-leukaemia' effect. However, due to the rarity of this disease, and the availability of mature B-cell antigens on the cell surface, few new cancer antigens have been identified in adult B-ALL that could act as targets to remove residual disease in first remission or provide alternative targets for escape variants if and when current immunotherapy strategies fail. We have used RT-PCR analysis, literature searches, antibody-specific profiling and gene expression microarray analysis to identify and prioritise antigens as novel targets for the treatment of adult B-ALL.
Collapse
Affiliation(s)
- Stephanie Jordaens
- Department of Biomedical Sciences, University of Hull, Cottingham Road, Hardy Building, Room 111, Hull, HU7 6RX, UK
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Leah Cooksey
- Department of Biomedical Sciences, University of Hull, Cottingham Road, Hardy Building, Room 111, Hull, HU7 6RX, UK
| | | | - Viggo Van Tendeloo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Evelien Smits
- Centre for Oncological Research, University of Antwerp, Antwerp, Belgium
| | - Ken I Mills
- Centre for Cancer Research and Cell Biology, Queens University Belfast, Lisburn Road, Belfast, UK
| | - Kim H Orchard
- Department of Haematology, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| | - Barbara-Ann Guinn
- Department of Biomedical Sciences, University of Hull, Cottingham Road, Hardy Building, Room 111, Hull, HU7 6RX, UK.
| |
Collapse
|
30
|
Fuchigami T, Ishikawa N, Nozaki I, Miyanari Y, Yoshida S, Yamauchi M, Soejima A, Haratake M, Nakayama M. Discovery of inner centromere protein-derived small peptides for cancer imaging and treatment targeting survivin. Cancer Sci 2020; 111:1357-1366. [PMID: 31991041 PMCID: PMC7156834 DOI: 10.1111/cas.14330] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 01/16/2020] [Accepted: 01/18/2020] [Indexed: 12/15/2022] Open
Abstract
Survivin belongs to the inhibitor of apoptosis protein family, which is consistently overexpressed in most cancer cells but rarely expressed in normal adult tissues. Therefore, the detection and inhibition of survivin are regarded as attractive strategies for cancer‐specific treatment. In this study, we designed and synthesized 7‐19 residues of inner centromere protein (INCENP)‐derived small peptides (INC peptides) as novel survivin‐targeting agents. The INC peptides showed binding affinity for the human survivin protein (Kd = 91.4‐255 nmol L−1); INC16‐22, which contains residues 16‐22 of INCENP, showed the highest affinity (91.4 nmol L−1). Confocal fluorescence imaging showed consistent colocalization of FITC‐INC16‐22 and survivin in cell lines. Nona‐arginine‐linked INC16‐22 (r9‐INC16‐22) rendered INC16‐22 cells penetrable and strongly inhibited cell growth of MIA PaCa‐2 cells (52% inhibition at 1.0 µmol L−1) and MDA‐MB‐231 cells (60% inhibition at 10 µmol L−1) as determined by MTT assays. The exposure of MIA PaCa‐2 cells to 40 µmol L−1 r9‐INC16‐22 apparently reduced the intracellular protein expression levels of survivin. However, cleaved caspase‐3 was significantly increased in cells treated with r9‐INC16‐22, even at 10 µmol L−1, compared to untreated cells. Flow cytometry revealed that r9‐INC16‐22 strongly induced apoptosis in MIA PaCa‐2 cells. These results indicate that the cytotoxic effects of r9‐INC16‐22 could be mediated mainly through the disruption of survivin‐dependent antiapoptotic functions and partly because of the direct degradation of the survivin protein. Our findings suggest that INC peptides can act as useful scaffolds for novel cancer imaging and anticancer agents.
Collapse
Affiliation(s)
- Takeshi Fuchigami
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Natsumi Ishikawa
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Iori Nozaki
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yusuke Miyanari
- Okazaki Institute for Integrative Bioscience, Okazaki, Japan
| | - Sakura Yoshida
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Motohiro Yamauchi
- Department of Radiation Biology and Protection, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Ayumi Soejima
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Mamoru Haratake
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Morio Nakayama
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
31
|
Brown M, Zhang W, Yan D, Kenath R, Le L, Wang H, Delitto D, Ostrov D, Robertson K, Liu C, Pham K. The role of survivin in the progression of pancreatic ductal adenocarcinoma (PDAC) and a novel survivin-targeted therapeutic for PDAC. PLoS One 2020; 15:e0226917. [PMID: 31929540 PMCID: PMC6957139 DOI: 10.1371/journal.pone.0226917] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 12/07/2019] [Indexed: 12/11/2022] Open
Abstract
Treating pancreatic ductal adenocarcinoma (PDAC) remains a major hurdle in the field of oncology. Less than half of patients respond to frontline chemotherapy and the pancreatic tumor microenvironment limits the efficacy of immunotherapeutic approaches. Targeted therapies could serve as effective treatments to enhance the clinical response rate. One potential therapeutic target is survivin, a protein that is normally expressed during embryonic and fetal development and has a critical impact on cell cycle control and apoptosis. In adulthood, survivin is not present in most normal adult cells, but is significantly re-expressed in tumor tissues. In PDAC, elevated survivin expression is correlated with treatment resistance and lower patient survival, although the underlying mechanisms of survivin's action in this type of cancer is poorly understood. Using patient derived xenografts of PDAC and their corresponding primary pancreatic cancer lines (PPCL-46 and PPCL-LM1) possessing increased expression of survivin, we aimed to evaluate the therapeutic response of a novel survivin inhibitor, UFSHR, with respect to survivin expression and the tumorigenic characteristics of PDAC. Cell viability and apoptosis analyses revealed that repressing survivin expression by UFSHR or YM155, a well-known inhibitor of survivin, in PPCLs effectively reduces cell proliferation by inducing apoptosis. Tumor cell migration was also hindered following treatment with YM155 and UFSHR. In addition, both survivin inhibitors, particularly UFSHR, effectively reduced progression of PPCL-46 and PPCL-LM1 tumors, when compared to the untreated cohort. Overall, this study provides solid evidence to support the critical role of survivin in PDAC progression and proposes a novel survivin inhibitor UFSHR that can become an alternative strategy for this type of cancer.
Collapse
Affiliation(s)
- Matthew Brown
- Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey, United States of America
| | - Wanbin Zhang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Deyue Yan
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Rajath Kenath
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Le Le
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - He Wang
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Daniel Delitto
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - David Ostrov
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Keith Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
- * E-mail: (KP); (CL)
| | - Kien Pham
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
- * E-mail: (KP); (CL)
| |
Collapse
|
32
|
Shen S, Dean DC, Yu Z, Hornicek F, Kan Q, Duan Z. Aberrant CDK9 expression within chordoma tissues and the therapeutic potential of a selective CDK9 inhibitor LDC000067. J Cancer 2020; 11:132-141. [PMID: 31892980 PMCID: PMC6930393 DOI: 10.7150/jca.35426] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/16/2019] [Indexed: 12/17/2022] Open
Abstract
Objectives: Chordomas are slow-growing malignancies that commonly affect vital neurological structures. These neoplasms are highly resistant to current chemotherapeutic regimens and often recur after surgical intervention. Therefore, there is an urgent need to identify molecular targets and more robust drugs to improve chordoma patient outcomes. It is well accepted that cyclin-dependent protein kinase 9 (CDK9) has tumorigenic roles in various cancers; however, the expression and significance of CDK9 in chordoma remains unknown. Methods: Expression of CDK9 in chordoma cell lines and tumor tissues was examined by Western blot and immunohistochemistry (IHC). The correlation between CDK9 expression in patient tissues and clinical prognosis was analyzed. The functional roles of CDK9 in chordoma were investigated after the addition of small interfering RNA (siRNA) and CDK9 inhibitor (LDC000067). Cell growth and proliferation were assessed with MTT and clonogenic assays. The effect of CDK9 inhibition on chordoma cells was further evaluated with a three-dimensional (3D) cell culture model which mimics the in vivo environment. Results: CDK9 was expressed in both chordoma cell lines and chordoma tissues. High- expression of CDK9 correlated with recurrence and poor outcomes for chordoma patients. CDK9 silencing with siRNA decreased growth and proliferation of chordoma cells and lowered levels of Mcl-1 and RNA polymerase II (RNAP II) phosphorylation. Pharmacological inhibition of CDK9 with the small molecular inhibitor LDC000067 reduced cell growth, supported apoptosis, suppressed cell colony formation in a clonogenic assay, and decreased spheroid growth in 3D culture. Conclusion: We demonstrate that CDK9 expression in chordoma correlates with patient outcome, and, when inhibited, chordoma cell growth and proliferation significantly decreases. Taken together, these results support CDK9 as an emerging potential target in chordoma therapy.
Collapse
Affiliation(s)
- Shen Shen
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.,Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, David Geffen School of Medicine at University of Los Angeles, Los Angeles, CA 90095, USA
| | - Dylan C Dean
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, David Geffen School of Medicine at University of Los Angeles, Los Angeles, CA 90095, USA
| | - Zujiang Yu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Francis Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, David Geffen School of Medicine at University of Los Angeles, Los Angeles, CA 90095, USA
| | - Quancheng Kan
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhenfeng Duan
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.,Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, David Geffen School of Medicine at University of Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
33
|
Huang YH, Yeh CT. Functional Compartmentalization of HSP60-Survivin Interaction between Mitochondria and Cytosol in Cancer Cells. Cells 2019; 9:cells9010023. [PMID: 31861751 PMCID: PMC7016642 DOI: 10.3390/cells9010023] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022] Open
Abstract
Heat shock protein 60 (HSP60) and survivin reside in both the cytosolic and mitochondrial compartments under physiological conditions. They can form HSP60-survivin complexes through protein–protein interactions. Their expression levels in cancer tissues are positively correlated and higher expression of either protein is associated with poor clinical prognosis. The subcellular location of HSP60-survivin complex in either the cytosol or mitochondria is cell type-dependent, while the biological significance of HSP60-survivin interaction remains elusive. Current knowledge indicates that the function of HSP60 partly rests on where HSP60-survivin interaction takes place. HSP60 has a pro-survival function when binding to survivin in the mitochondria through interacting with other factors such as CCAR2 and p53. In response to cell death signals, mitochondrial survivin functions through preventing procaspase activation. Degradation of cytosolic survivin leads to the loss of mitochondrial membrane potential and aberrant mitosis processes. On the other hand, HSP60 release from mitochondria to cytosol upon death stimuli might exert a pro-death function, either through stabilizing Bax, enhancing procaspase-3 activation, or increasing protein ubiquitination. Combining the knowledge of mitochondrial HSP60-survivin complex function, cytosolic survivin degradation effect, and pro-death function upon mitochondria release of HSP60, a hypothetical scenario for HSP60-survivin shuttling upon death stimuli is proposed.
Collapse
|
34
|
Sun X, Angelastro JM, Merino D, Zhou Q, Siegelin MD, Greene LA. Dominant-negative ATF5 rapidly depletes survivin in tumor cells. Cell Death Dis 2019; 10:709. [PMID: 31551409 PMCID: PMC6760124 DOI: 10.1038/s41419-019-1872-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 07/22/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022]
Abstract
Survivin (BIRC5, product of the BIRC5 gene) is highly expressed in many tumor types and has been widely identified as a potential target for cancer therapy. However, effective anti-survivin drugs remain to be developed. Here we report that both vector-delivered and cell-penetrating dominant-negative (dn) forms of the transcription factor ATF5 that promote selective death of cancer cells in vitro and in vivo cause survivin depletion in tumor cell lines of varying origins. dn-ATF5 decreases levels of both survivin mRNA and protein. The depletion of survivin protein appears to be driven at least in part by enhanced proteasomal turnover and depletion of the deubiquitinase USP9X. Survivin loss is rapid and precedes the onset of cell death triggered by dn-ATF5. Although survivin downregulation is sufficient to drive tumor cell death, survivin over-expression does not rescue cancer cells from dn-ATF5-promoted apoptosis. This indicates that dn-ATF5 kills malignant cells by multiple mechanisms that include, but are not limited to, survivin depletion. Cell-penetrating forms of dn-ATF5 are currently being developed for potential therapeutic use and the present findings suggest that they may pose an advantage over treatments that target only survivin.
Collapse
Affiliation(s)
- Xiaotian Sun
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - James M Angelastro
- Department of Molecular Biosciences, University of California, Davis School of Veterinary Medicine, Davis, CA, 95616, USA
| | - David Merino
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA.,CMI Strategies, Boulogne-Billancourt, 80 rue Gallieni, cedex, France
| | - Qing Zhou
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - Lloyd A Greene
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
35
|
Wang L, En H, Yang L, Zhang Y, Sun B, Gao J. miR-596 suppresses the expression of Survivin and enhances the sensitivity of osteosarcoma cells to the molecular targeting agent anlotinib. Onco Targets Ther 2019; 12:6825-6838. [PMID: 31686840 PMCID: PMC6709039 DOI: 10.2147/ott.s215145] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/31/2019] [Indexed: 12/18/2022] Open
Abstract
Background Osteosarcoma (OSA), the most common primary bone malignancy, is characterized by a wide spectrum of complicated pathologies and frequent distal metastasis and causes death in adolescents and young adults worldwide. Antitumor drug treatment strategies include various cytotoxic chemotherapy drugs, while molecular targeted therapy for OSA is currently less used. The present work revealed the role played by the miR-596/Survivin axis in affecting the sensitivity of OSA cells to anlotinib, a novel molecular targeting agent. Methods By virtual screening, we found that miR-596 might target Survivin by using an online tool (miRDB). RNA levels of miR-596 and Survivin in clinical specimens were examined with qPCR. The effect of miR-596 on anlotinib’s antitumor effect was examined with MTT experiments, the subcutaneous tumor model, or the intramuscular tumor model. Results Overexpression of miR-596 via lentiviral particles repressed the protein level of Survivin in U2OS cells. Transfection of miR-596 enhanced the antitumor effect of anlotinib on U2OS cells or five cell lines derived from OSA patients. Conclusion miR-596 targets Survivin and enhances the antitumor effect of anlotinib on OSA cells.
Collapse
Affiliation(s)
- Leisheng Wang
- Department of Orthopedics, Yantaishan Hospital, Yantai, Shandong Province 264000, People's Republic of China
| | - He En
- Department of Outpatient, The 81st Group Army Hospital of Chinese People's Liberation Army, Zhangjiakou City, Hebei Province, People's Republic of China
| | - Lei Yang
- Department of Outpatient, The 80th Group Army Hospital of Chinese People's Liberation Army (formerly the 89th Hospital of the People's Liberation Army), Weifang City, Shandong Province, People's Republic of China
| | - Yanbing Zhang
- Department of Outpatient, The 81st Group Army Hospital of Chinese People's Liberation Army, Zhangjiakou City, Hebei Province, People's Republic of China
| | - Baisheng Sun
- Department of Emergency, The Fifth Medical Center of the General Hospital of the Chinese People's Liberation Army (formerly the 307th Hospital of the People's Liberation Army), Beijing 100071, People's Republic of China
| | - Jianjiang Gao
- Department of Emergency, Haiyang People's Hospital, Haiyang, Shandong 265100, People's Republic of China
| |
Collapse
|
36
|
Moazeni-Roodi A, Ghavami S, Hashemi M. Survivin rs9904341 polymorphism significantly increased the risk of cancer: evidence from an updated meta-analysis of case-control studies. Int J Clin Oncol 2019; 24:335-349. [PMID: 30747314 DOI: 10.1007/s10147-019-01408-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 01/27/2019] [Indexed: 12/11/2022]
Abstract
AIMS Survivin, a member of inhibitor of apoptosis protein family, is involved in the regulation of cell cycle and apoptosis. Several studies inspected the association between survivin polymorphisms and the risk of various cancers, but the findings remain controversial. We conducted a meta-analysis intending to certify the association between survivin polymorphisms and cancer risk. METHODS All analyses were achieved using RevMan 5.3 software and STATA 14.1 software. Eligible studies were collected by comprehensive literature searching Web of Science, PubMed, Scopus, and Google scholar databases. Pooled estimates of odds ratios (ORs) with 95% confidence intervals (CIs) were used to assess the overall impact of survivin polymorphisms on cancer risk. RESULTS The overall analysis indicates that survivin rs9904341 polymorphism significantly increased the risk of cancer in homozygous codominant (OR 1.41, 95% CI 1.19-1.68, p = 0.0001, CC vs GG), dominant (OR 1.22, 95% CI 1.07-1.40, p = 0.003, CG+CC vs GG), recessive (OR 1.34, 95% CI 1.18-1.52, p < 0.0001, CC vs CG+GG), and allele (OR 1.20, 95% CI 1.09-1.31, p = 0.0001, C vs G) inheritance models tested. Stratified based on ethnicity revealed that rs9904341 variant significantly increased the risk of cancer in the Asian population. The findings did not support an association between rs1042489, rs2071214, rs8073069, and rs17878467 polymorphisms and risk of cancer. CONCLUSIONS The current study suggests that the survivin rs9904341 polymorphism may be associated with the risk of cancer either overall or in the Asian population. However, further larger and well-designed studies are warranted to evaluate this association in detail.
Collapse
Affiliation(s)
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Research Institute in Oncology and Hematology, CancerCare Manitoba, University of Manitoba, Winnipeg, Canada
| | - Mohammad Hashemi
- Genetics of Non-communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| |
Collapse
|
37
|
Zhao Z, Huang R, Cai H, Liu B, Zeng Y, Kuang A. Radionuclide imaging and therapy in malignant melanoma after survivin promoter-directed sodium iodide symporter gene transfer in vitro and in vivo. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:613-618. [PMID: 31933867 PMCID: PMC6945074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 12/26/2018] [Indexed: 06/10/2023]
Abstract
This study aimed to develop a gene expression targeting method specific for the imaging and therapy of malignant melanoma A375 cells using the sodium iodide symporter gene under control of the survivin promoter (Ad-Sur-NIS). When compared to control Ad-Sur-GFP-treated cells, Ad-Sur-NIS resulted in significantly higher iodide uptake in all 50, 100, or 150 MOIs examined cells (P<0.001). In vitro clonogenic assay showed the inhibition rates induced by 131I were 94.8±12.4% in Ad-Sur-NIS, which was significantly higher than that in Ad-Sur-GFP infected cells (12.5±2.3%, P<0.001) or untreated cells (11.1±1.8%, P<0.001). In biodistribution studies, the tumor-to-muscle ratio in Ad-Sur-NIS infected tumors was higher than that in Ad-Sur-GFP infected tumors (16.34±4.43 vs 1.44±0.39, P<0.001). Moreover, mice that received the injection of Ad-Sur-NIS and 131I showed a significant delay in tumor growth. Taken together, Ad-Sur-NIS expresses functional NIS, resulting in intracellular accumulation of radionuclide in malignant melanoma A375 cells in vitro and in vivo.
Collapse
Affiliation(s)
- Zhen Zhao
- Department of Nuclear Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Rui Huang
- Department of Nuclear Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Huawei Cai
- Department of Nuclear Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Bin Liu
- Department of Nuclear Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Yu Zeng
- Department of Nuclear Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Anren Kuang
- Department of Nuclear Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| |
Collapse
|