1
|
Fan Y, Pan Y, Jia L, Gu S, Liu B, Mei Z, Lv C, Huang H, Zhu G, Deng Q. BIRC5 facilitates cisplatin-chemoresistance in a m 6A-dependent manner in ovarian cancer. Cancer Med 2024; 13:e6811. [PMID: 38112021 PMCID: PMC10807614 DOI: 10.1002/cam4.6811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 08/26/2023] [Accepted: 09/26/2023] [Indexed: 12/20/2023] Open
Abstract
Cisplatin-based chemotherapy is the standard treatment for metastatic ovarian cancer (OC). However, chemoresistance continues to pose significant clinical challenges. Recent research has highlighted the baculoviral inhibitor of the apoptosis protein repeat-containing 5 (BIRC5) as a member of the inhibitor of the apoptosis protein (IAP) family. Notably, BIRC5, which has robust anti-apoptotic capabilities, is overexpressed in numerous cancers. Its dysfunction has been linked to challenges in cancer treatment. Yet, the role of BIRC5 in the chemoresistance of OC remains elusive. In our present study, we observed an upregulation of BIRC5 in cisplatin-resistant cell lines. This upregulation was associated with enhanced chemoresistance, which was diminished when the expression of BIRC5 was silenced. Intriguingly, BIRC5 exhibited a high number of N6-methyladenosine (m6A) binding sites. The modification of m6A was found to enhance the expression of BIRC5 by recognizing and binding to the 3'-UTR of mRNA. Additionally, the insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) was shown to stabilize BIRC5 mRNA, synergizing with METTL3 and intensifying chemoresistance. Supporting these in vitro findings, our in vivo experiments revealed that tumors were significantly smaller in size and volume when BIRC5 was silenced. This reduction was notably counteracted by co-silencing BIRC5 and overexpressing IGF2BP1. Our results underscored the pivotal role of BIRC5 in chemoresistance. The regulation of its expression and the stability of its mRNA were influenced by m6A modifications involving both METTL3 and IGF2BP1. These insights presented BIRC5 as a promising potential therapeutic target for addressing cisplatin resistance in OC.
Collapse
Affiliation(s)
- Yadan Fan
- Department of GynecologyThe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Yinglian Pan
- Department of OncologyThe First Affiliated Hospital of Hainan Medical CollegeHaikouChina
| | - Liping Jia
- Department of GynecologyThe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Shuzhen Gu
- Department of GynecologyThe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Binxin Liu
- Department of GynecologyThe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Ziman Mei
- Department of GynecologyThe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Chunyan Lv
- Department of GynecologyThe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Haohao Huang
- Department of NeurosurgeryGeneral Hospital of Central Theater Command of Chinese People's Liberation ArmyWuhanChina
| | - Genhai Zhu
- Department of GynecologyHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Qingchun Deng
- Department of GynecologyThe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
| |
Collapse
|
2
|
Tampouratzi E, Asonitis K, Katsantonis J, Talaiporou K, Sfaelos K. Preoperative and postoperative evaluation of survivin in peripheral blood of patients with non-melanoma skin cancers. A pilot study. J DERMATOL TREAT 2023; 34:2147390. [PMID: 36369849 DOI: 10.1080/09546634.2022.2147390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
| | - Konstantinos Asonitis
- Department of Internal Medicine, Réseau Hospitalier Neuchâtelois, Neuchâtel, Switzerland
| | - John Katsantonis
- Department of Dermatology and Venereology, Tzaneio General Hospital, Piraeus, Greece
| | - Kyriakos Talaiporou
- Department of Business Administration, University of Western Attica, Athens, Greece
| | - Konstantinos Sfaelos
- Department of Skin & Venereal Diseases, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| |
Collapse
|
3
|
Li C, Zhu M, Lu Q, Yu B, Yang Y, Li B, Peng Z, Li Z. Oncolytic adenovirus-mediated dual knockdown of survivin and OCT4 improves therapeutic efficacy in esophageal cancer. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:193. [PMID: 37007541 PMCID: PMC10061493 DOI: 10.21037/atm-22-4499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 01/19/2023] [Indexed: 03/08/2023]
Abstract
Background Survivin and octamer-binding transcription factor 4 (OCT4) are reportedly up-regulated in esophageal cancer (EC) and have been correlated with high tumor proliferative activity and poor prognosis. Oncolytic viruses encoding specific transgenes have been considered as therapeutic methods to increase therapeutic efficacy in a variety of solid tumors. Methods In this study, an oncolytic adenovirus carrying short hairpin RNA (shRNA) of survivin (shSRVN) and OCT4 (shOCT4) was constructed to achieve dual knockdown of survivin and OCT4 and to explore the potential effect of the oncolytic adenovirus in EC. Results The oncolytic adenovirus replicated abundantly in human EC cells, with the replication multiplying by up to 192,085 and 620,055 times in esophageal carcinoma (Eca)-109 cells transfected with purified and completed recombinant adenoviruses called AdSProE1a-dual shRNA (shSRVN + shOCT4) and TE1 cells transfected with AdSProE1a-survivin shRNA (shSRVN) 96 hours after infection, respectively. The shRNAs targeting survivin and OCT4 significantly downregulated the expression levels of survivin and OCT4 in cells, thereby inhibiting the proliferative activity of cancer cells. Furthermore, E-cadherin and vimentin, which are both considered epithelial mesenchymal transition (EMT) markers, were found to be upregulated and downregulated, respectively, in cancer cells after exposure to the viral infection. The interference of survivin and OCT4 also contributed to cell cycle arrest and apoptosis, the half maximal inhibitory concentrations (IC50s) of oncolytic adenovirus loaded with AdSProE1a-shSRVN + shOCT4 in the Eca109 cells and the TE1 cells were 0.7271 and 0.1032 pfu/mL, respectively. Xenograft experiments in vivo showed that oncolytic adenovirus-mediated dual knockdown of survivin and OCT4 effectively inhibited the growth of xenografts and induced cancer cell apoptosis. We concluded that therapies targeting survivin and OCT4 have great potential for improving the therapeutic efficacy in EC. Conclusions The dual target design strategy ensured the efficacy and safety of the treatment system and provided a novel and effective adjuvant target therapy for EC.
Collapse
Affiliation(s)
- Chunguang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Maoling Zhu
- Department of Gastroenterology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qijue Lu
- Department of Thoracic Surgery, Naval Medical Center, Naval Medical University, Shanghai, China
| | - Boyao Yu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhangxiao Peng
- Department of Molecular Oncology, Eastern Hepatobiliary Surgery Hospital & National Centre for Liver Cancer, Naval Medical University, Shanghai, China
| | - Zhigang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
4
|
Prognostic impact of lymphovascular and perineural invasion in squamous cell carcinoma of the tongue. Sci Rep 2023; 13:3828. [PMID: 36882521 PMCID: PMC9992656 DOI: 10.1038/s41598-023-30939-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
This study aimed to investigate the prognostic impact of lymphovascular and perineural invasions in patients with squamous cell carcinoma of the tongue who received surgery-based treatment at our institution between January 2013 and December 2020. Patients were divided into four groups based on the presence of perineural (P-/P +) and lymphovascular invasions (V-/V +): P-V-, P-V + , P + V-, and P + V + . Log-rank and Cox proportional hazard models were used to evaluate the association between perineural /lymphovascular invasion and overall survival (OS). Altogether, 127 patients were included, and 95 (74.8%), 8 (6.3%), 18 (14.2%), and 6 (4.7%) cases were classified as P-V-, P-V + , P + V-, and P + V + , respectively. Pathologic N stage (pN stage), tumor stage, histological grade, lymphovascular invasion, perineural invasion, and postoperative radiotherapy were significantly associated with OS (p < 0.05). OS was significantly different among the four groups (p < 0.05). Significant between-group differences in OS were detected for node-positive (p < 0.05) and stage III-IV (p < 0.05) cases. OS was the worst in the P + V + group. Lymphovascular and perineural invasions are independent negative prognostic factors for squamous cell carcinoma of the tongue. Patients with lymphovascular and/or perineural invasion may have significantly poorer overall survival than those without neurovascular involvement.
Collapse
|
5
|
Lnc Tmem235 promotes repair of early steroid-induced osteonecrosis of the femoral head by inhibiting hypoxia-induced apoptosis of BMSCs. Exp Mol Med 2022; 54:1991-2006. [PMID: 36380019 PMCID: PMC9723185 DOI: 10.1038/s12276-022-00875-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 03/24/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) have been used in the treatment of early steroid-induced osteonecrosis of the femoral head (SONFH). However, the hypoxic microenvironment in the osteonecrotic area leads to hypoxia-induced apoptosis of transplanted BMSCs, which limits their efficacy. Therefore, approaches that inhibit hypoxia-induced apoptosis of BMSCs are promising for augmenting the efficacy of BMSC transplantation. Our present study found that under hypoxia, the expression of the long noncoding RNA (Lnc) transmembrane protein 235 (Tmem235) was downregulated, the expression of Bcl-2-associated X protein was upregulated, the expression of B-cell lymphoma-2 protein was downregulated, and the apoptotic rate of BMSCs was over 70%. However, overexpression of Lnc Tmem235 reversed hypoxia-induced apoptosis of BMSCs and promoted their survival. These results demonstrated that Lnc Tmem235 effectively inhibited hypoxia-induced apoptosis of BMSCs. Mechanistically, we found that Lnc Tmem235 exhibited competitive binding to miR-34a-3p compared with BIRC5 mRNA, which is an inhibitor of apoptosis; this competitive binding relieved the silencing effect of miR-34a-3p on BIRC5 mRNA to ultimately inhibit hypoxia-induced apoptosis of BMSCs by promoting the expression of BIRC5. Furthermore, we cocultured BMSCs overexpressing Lnc Tmem235 with xenogeneic antigen-extracted cancellous bone to construct tissue-engineered bone to repair a model of early SONFH in vivo. The results showed that overexpression of Lnc Tmem235 effectively reduced apoptosis of BMSCs in the hypoxic microenvironment of osteonecrosis and improved the effect of BMSC transplantation. Taken together, our findings show that Lnc Tmem235 inhibited hypoxia-induced apoptosis of BMSCs by regulating the miR-34a-3p/BIRC5 axis, thus improving the transplantation efficacy of BMSCs for treating early SONFH.
Collapse
|
6
|
Chandrasekaran AP, Tyagi A, Poondla N, Sarodaya N, Karapurkar JK, Kaushal K, Park CH, Hong SH, Kim KS, Ramakrishna S. Dual role of deubiquitinating enzyme USP19 regulates mitotic progression and tumorigenesis by stabilizing survivin. Mol Ther 2022; 30:3414-3429. [PMID: 35918893 PMCID: PMC9637645 DOI: 10.1016/j.ymthe.2022.07.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 06/09/2022] [Accepted: 07/30/2022] [Indexed: 11/22/2022] Open
Abstract
Survivin is a component of the chromosomal passenger complex, which includes Aurora B, INCENP, and Borealin, and is required for chromosome segregation and cytokinesis. We performed a genome-wide screen of deubiquitinating enzymes for survivin. For the first time, we report that USP19 has a dual role in the modulation of mitosis and tumorigenesis by regulating survivin expression. Our results found that USP19 stabilizes and interacts with survivin in HCT116 cells. USP19 deubiquitinates survivin protein and extends its half-life. We also found that USP19 functions as a mitotic regulator by controlling the downstream signaling of survivin protein. Targeted genome knockout verified that USP19 depletion leads to several mitotic defects, including cytokinesis failure. In addition, USP19 depletion results in significant enrichment of apoptosis and reduces the growth of tumors in the mouse xenograft. We envision that simultaneous targeting of USP19 and survivin in oncologic drug development would increase therapeutic value and minimize redundancy.
Collapse
Affiliation(s)
- Arun Pandian Chandrasekaran
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, 222 Wangsimni-ro, Seongdong, Seoul 04763, South Korea
| | - Apoorvi Tyagi
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, 222 Wangsimni-ro, Seongdong, Seoul 04763, South Korea
| | - Naresh Poondla
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, 222 Wangsimni-ro, Seongdong, Seoul 04763, South Korea
| | - Neha Sarodaya
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, 222 Wangsimni-ro, Seongdong, Seoul 04763, South Korea
| | - Janardhan Keshav Karapurkar
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, 222 Wangsimni-ro, Seongdong, Seoul 04763, South Korea
| | - Kamini Kaushal
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, 222 Wangsimni-ro, Seongdong, Seoul 04763, South Korea
| | - Chang-Hwan Park
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, 222 Wangsimni-ro, Seongdong, Seoul 04763, South Korea; College of Medicine, Hanyang University, Seoul 04763, South Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, South Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, 222 Wangsimni-ro, Seongdong, Seoul 04763, South Korea; College of Medicine, Hanyang University, Seoul 04763, South Korea.
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, 222 Wangsimni-ro, Seongdong, Seoul 04763, South Korea; College of Medicine, Hanyang University, Seoul 04763, South Korea.
| |
Collapse
|
7
|
Cell death mechanisms in head and neck cancer cells in response to low and high-LET radiation. Expert Rev Mol Med 2022. [DOI: 10.1017/erm.2021.31] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AbstractHead and neck squamous cell carcinoma (HNSCC) is a common malignancy that develops in or around the throat, larynx, nose, sinuses and mouth, and is mostly treated with a combination of chemo- and radiotherapy (RT). The main goal of RT is to kill enough of the cancer cell population, whilst preserving the surrounding normal and healthy tissue. The mechanisms by which conventional photon RT achieves this have been extensively studied over several decades, but little is known about the cell death pathways that are activated in response to RT of increasing linear energy transfer (LET), including proton beam therapy and heavy ions. Here, we provide an up-to-date review on the observed radiobiological effects of low- versus high-LET RT in HNSCC cell models, particularly in the context of specific cell death mechanisms, including apoptosis, necrosis, autophagy, senescence and mitotic death. We also detail some of the current therapeutic strategies targeting cell death pathways that have been investigated to enhance the radiosensitivity of HNSCC cells in response to RT, including those that may present with clinical opportunities for eventual patient benefit.
Collapse
|
8
|
Chintala L, Hussain KA, Kodangal S, Lingam AS, Dantu R, Nagabhyrava P. Expression of survivin in oral leukoplakia and oral lichen planus. Minerva Dent Oral Sci 2021; 71:125-130. [PMID: 34672483 DOI: 10.23736/s2724-6329.21.04578-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Up to one third (3-33%) oral squamous cell carcinomas develop from potentially malignant lesions. Most common potentially malignant lesions (PML) are Leukoplakia and Lichen planus. It is very important to diagnose PML at early stage to prevent malignant transformation of lesion. Survivin is a smallest member of the Inhibitor of Apoptosis (IAP) family of proteins. Survivin plays an important role in apoptosis regulation. High expression of survivin is an early event during oral carcinogenesis and it acts as a tool for the identification of precancerous lesions at higher risk of progression into invasive carcinoma. The aim of the study was to evaluate the expression of Survivin in oral leukoplakia, oral lichen planus. METHODS AND MATERIAL 15 patients with oral lichen planus, 15 patients of oral leukoplakia were selected as subjects for the present study and 15 patients with normal oral mucosa were selected as controls and all were evaluated for expression of survivin. All sections were H & E stained and were studied immunohistochemically for the expression of surviving. RESULTS Expression of survivin was evaluated based on percent of cells expressing survivin and grading was given according to that. Significant survivin expression was detected in oral leukoplakia, oral lichen planus, (P < 0.001). CONCLUSIONS Expression of survivin in potentially malignant lesions indicate potential risk of malignant transformation.
Collapse
Affiliation(s)
- Lalitha Chintala
- Department of Oral Medicine and Radiology, Malla Reddy Institute of Dental Sciences, Hyderabad, Telangana, India
| | | | | | - Amara S Lingam
- Department of Surgical and Diagnostic Sciences, Dar Al Uloom University, Riyadh, Saudi Arabia -
| | - Raghunath Dantu
- Department of Oral Medicine and Radiology, Malla Reddy Institute of Dental Sciences, Hyderabad, Telangana, India
| | | |
Collapse
|
9
|
Natural Polymers for the Maintenance of Oral Health: Review of Recent Advances and Perspectives. Int J Mol Sci 2021; 22:ijms221910337. [PMID: 34638678 PMCID: PMC8508910 DOI: 10.3390/ijms221910337] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 12/14/2022] Open
Abstract
The success of modern dental treatment is strongly dependent on the materials used both temporarily and permanently. Among all dental materials, polymers are a very important class with a wide spectrum of applications. This review aims to provide a state-of-the-art overview of the recent advances in the field of natural polymers used to maintain or restore oral health. It focuses on the properties of the most common proteins and polysaccharides of natural origin in terms of meeting the specific biological requirements in the increasingly demanding field of modern dentistry. The use of naturally derived polymers in different dental specialties for preventive and therapeutic purposes has been discussed. The major fields of application cover caries and the management of periodontal diseases, the fabrication of membranes and scaffolds for the regeneration of dental structures, the manufacturing of oral appliances and dentures as well as providing systems for oral drug delivery. This paper also includes a comparative characteristic of natural and synthetic dental polymers. Finally, the current review highlights new perspectives, possible future advancements, as well as challenges that may be encountered by researchers in the field of dental applications of polymers of natural origin.
Collapse
|
10
|
Carbonic Anhydrase IX and Survivin in Colorectal Adenocarcinoma Cells: Slovakian Population Study. BIOLOGY 2021; 10:biology10090872. [PMID: 34571749 PMCID: PMC8466885 DOI: 10.3390/biology10090872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/20/2021] [Accepted: 08/28/2021] [Indexed: 11/17/2022]
Abstract
Simple Summary This retrospective study (Slovakian population study) brings information about immunohistochemical detection of CAIX and survivin in 74 samples of human colorectal adenocarcinoma and comparison their expression with expression in healthy colon tissue. Our results show that all of samples with healthy colon tissue were CAIX and survivin-negative and there is no statistically significant dependence of these proteins and the chosen clinicopathological parameters. These findings demonstrate that detection of these proteins could be useful for tumor diagnostic and prognostic and CAIX and survivin could represent independent negative prognostic markers of colorectal cancer. Abstract The aim of this study was to detect carbonic anhydrase IX (CAIX) and survivin in the colorectal adenocarcinoma cells of the Slovakian population. We used an indirect three-step immunohistochemical method with DAB staining for the localization of the proteins and investigation their expression. We compared their expression with expression in healthy colorectal tissue. In 74 tissues of colorectal adenocarcinomas, 42% showed CAIX positivity and 20% showed survivin positivity. Brown membrane immunostaining was visible in CAIX-positive tumors. Survivin-positive tumors had strong brown cytoplasmic immunostaining. Co-expression of both proteins was present in five specimens (7%). The samples of normal colorectal tissue (without carcinoma) were CAIX-negative and survivin-negative. We also applied the Chi-squared test for evaluation statistically significant association between the expression of proteins and selected clinical and histopathological parameters. We did not find any statistically significant correlations between CAIX or survivin expression and sex of patients, the grade of the tumor, nodal status and presence of metastasis (p > 0.05). The fact that all samples of normal colorectal tissue were CAIX- and survivin-negative could lead to the possibility of using these two proteins as potential tumor diagnostic markers. On the basic of the available publications and data, we suggest that CAIX and survivin could be negative independent prognostic markers of colorectal cancer, which could affect response to therapy.
Collapse
|
11
|
Paterniti I, Scuderi SA, Casili G, Lanza M, Mare M, Giuffrida R, Colarossi C, Portelli M, Cuzzocrea S, Esposito E. Poly (ADP-Ribose) Polymerase Inhibitor, ABT888, Improved Cisplatin Effect in Human Oral Cell Carcinoma. Biomedicines 2021; 9:biomedicines9070771. [PMID: 34356835 PMCID: PMC8301366 DOI: 10.3390/biomedicines9070771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/16/2021] [Accepted: 06/28/2021] [Indexed: 11/16/2022] Open
Abstract
Cisplatin is one of the chemotherapeutic drugs used for the management of oral carcinoma, in which combined therapies are estimated to exert superior therapeutic efficacy compared with monotherapy. It is known that poly(ADP-ribosyl)ation is implicated in a multiplicity of cellular activities, such as DNA repair and cell death. Based on these, PARP inhibitors are used for the treatment of cancers; however, the capacity of PARP inhibitors associated to anti-cancer drugs have not been completely assessed in oral carcinoma. Here, we evaluated the effects of PARPi veliparib (ABT888) in combination with cisplatin on the survival of three human oral cancer cell lines HSC-2, Ca9-22 and CAL27 and we observed the effects of ABT888 alone or in combination with cisplatin on apoptosis and DNA damage repair mechanism. The results obtained showed that ABT888 induces a cytotoxicity effect on cell viability increasing the apoptotic pathway as well as DNA strand break; moreover, our results displayed the effects with cisplatin in a dose-dependent manner. Therefore, our results indicate PARP inhibitors as adjuvants for therapeutic strategy of oral cancer.
Collapse
Affiliation(s)
- Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, ME, Italy; (I.P.); (S.A.S.); (G.C.); (M.L.); (S.C.)
| | - Sarah Adriana Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, ME, Italy; (I.P.); (S.A.S.); (G.C.); (M.L.); (S.C.)
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, ME, Italy; (I.P.); (S.A.S.); (G.C.); (M.L.); (S.C.)
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, ME, Italy; (I.P.); (S.A.S.); (G.C.); (M.L.); (S.C.)
| | - Marzia Mare
- Istituto Oncologico del Mediterraneo, via Penninazzo 7, 95029 Viagrande, CT, Italy; (M.M.); (C.C.)
- IOM Ricerca Srl, via Penninazzo 11, 95029 Viagrande, CT, Italy;
| | | | - Cristina Colarossi
- Istituto Oncologico del Mediterraneo, via Penninazzo 7, 95029 Viagrande, CT, Italy; (M.M.); (C.C.)
| | - Marco Portelli
- Department of Biomedical and Dental Science, Morphological and Functional Images, University of Messina, via Consolare Valeria, 98125 Messina, ME, Italy;
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, ME, Italy; (I.P.); (S.A.S.); (G.C.); (M.L.); (S.C.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, ME, Italy; (I.P.); (S.A.S.); (G.C.); (M.L.); (S.C.)
- Correspondence: ; Tel.: +39-090-676-5208
| |
Collapse
|
12
|
Che H, Che Y, Zhang Z, Lu Q. Long Non-Coding RNA LINC01929 Accelerates Progression of Oral Squamous Cell Carcinoma by Targeting the miR-137-3p/FOXC1 Axis. Front Oncol 2021; 11:657876. [PMID: 33968763 PMCID: PMC8097103 DOI: 10.3389/fonc.2021.657876] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 03/22/2021] [Indexed: 12/18/2022] Open
Abstract
Recently, additional long noncoding RNAs (lncRNAs) have been identified and their possible roles were investigated in a variety of human tumors. One of these lncRNAs, LINC01929, promoted the progression of some cancers, whereas its expression and biological function in human oral squamous cell carcinoma (OSCC) remains still mostly uncertain. The LINC01929 expression in OSCC tissues or cell lines was identified via quantitative real-time polymerase chain reaction. The cell counting kit-8, transwell migration, wound-healing, and flow cytometry assays were utilized to characterize the functions of LINC01929 in OSCC cells. The interactive relationships between LINC01929 and miR-137-3p, miR-137-3p and Forkhead box C1 (FOXC1) were investigated by the dual-luciferase activity assay. Our findings demonstrated that LINC01929 was highly expressed in OSCC tissue samples and cell lines, whereas miR-137-3p expression was downregulated. LINC01929 acted as a carcinogenic lncRNA with accelerated OSCC cell proliferation, migration and invasion, and suppression of apoptosis. We further indicated that LINC01929 facilitated tumor growth in xenograft mouse models. Mechanistically, LINC01929 acted as a sponge for miR-137-3p to elevate FOXC1 expression, which is the target of miR-137-3p. In addition, downregulated miR-137-3p expression rescued the suppressive behaviors of LINC01929 knockdown on the biological behaviors of OSCC cells. Taken together, LINC01929 functioned as a tumor-promoting lncRNA via the miR-137-3p/FOXC1 axis in OSCC, suggesting novel targets for OSCC therapy.
Collapse
Affiliation(s)
- Hongze Che
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yanhai Che
- Department of Science and Education, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhimin Zhang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Qing Lu
- Department of General Dentistry, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
13
|
Zhou LQ, Hu Y, Xiao HJ. The prognostic significance of survivin expression in patients with HNSCC: a systematic review and meta-analysis. BMC Cancer 2021; 21:424. [PMID: 33863308 PMCID: PMC8052826 DOI: 10.1186/s12885-021-08170-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 04/07/2021] [Indexed: 12/16/2022] Open
Abstract
Background Survivin has been recently identified as a promising novel therapeutic target and prognostic marker in different types of cancer. Here we conducted a comprehensive meta-analysis to better clarify they the precise prognostic and diagnostic value of survivin in head and neck squamous cell carcinoma (HNSCC). Methods Database of PubMed (Medline), Embase, and Web of Science were systematically searched for related published literature up to September 2020. Pooled hazards ratios (HR) and related 95% confidence intervals (CI) were used to estimate the association of survivin expression and survival outcomes in HNSCC patients. Results Twenty eight studies with 4891 patients were finally included in this meta-analysis, the pooled analysis indicated that the survivin expression was significantly correlated with poorer overall survival (OS) (HR, 2.02; 95% CI, 1.65–2.47, P < 0.001), and poorer disease-free survival (DFS)/ disease-specific survival (DSS) (HR = 2.03, 95%CI: 1.64–2.52, P < 0.001; HR = 1.92, 95%CI: 1.41–2.60, P < 0.001, receptively). Similar results were observed in subgroup analysis stratified by different cancer types, such as laryngeal squamous cell carcinoma (LSCC) (HR = 1.35, 95%CI: 1.05–1.74, P < 0.001), oral squamous cell carcinomas (OSCC) (HR = 2.45, 95%CI: 1.89–3.17, P < 0.001), nasopharyngeal carcinoma (NPC) (HR = 2.53, 95%CI: 1.76–3.62, P < 0.001) and HNSCC (HR = 1.52, 95%CI: 1.25–1.86, P < 0.001). Furthermore, ethnicity-stratified analysis indicated that survivin was significantly associated with poorer OS among both Asian and Non- Asian HNSCC patients (HR = 2.16, 95%CI: 1.76–2.66; HR = 1.56, 95%CI: 1.33–1.82, respectively). Conclusions Our results suggested that survivin is predictors of worse prognosis in HNSCC patients. Hence, survivin is a potential therapeutic target for HNSCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08170-3.
Collapse
Affiliation(s)
- Liu-Qing Zhou
- Department of Otorhinolaryngology, Union Hospital, Ongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yao Hu
- Department of Otorhinolaryngology, The Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Hong-Jun Xiao
- Department of Otorhinolaryngology, Union Hospital, Ongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
14
|
Lymphovascular invasion as a prognostic tool for oral squamous cell carcinoma: a comprehensive review. Int J Oral Maxillofac Surg 2021; 51:1-9. [PMID: 33814227 DOI: 10.1016/j.ijom.2021.03.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 02/01/2021] [Accepted: 03/11/2021] [Indexed: 12/20/2022]
Abstract
Oral cancer is the most common malignancy of the head and neck region, characterized by a poor prognosis. Novel prognostic markers are needed to better stratify these patients. Lymphovascular invasion (LVI) has been included in the eighth edition of the AJCC Cancer Staging Manual as an additional prognostic factor, but its influence on the recurrence risk and lymph node metastasis is relatively understudied. This is a comprehensive review of the literature on the clinical and prognostic role of LVI in oral cancer. A relevant search of the PubMed, Scopus, and Web of Science databases yielded 29 articles that satisfied the inclusion criteria. Findings indicated that LVI is an independent negative prognostic factor in oral cancer patients and appears to be associated with cervical lymph node metastasis and loco-regional recurrence. Notably, in oral tongue cancer, survival outcomes progressively worsen when LVI is associated with other adverse pathological features, especially in the early stages. Therefore, these patients could benefit from elective neck dissection and/or adjuvant therapy. The high variability of LVI prevalence hinders the comparison of literature results. Several methodological limitations were found to be present in the collected articles, including the lack of a rigorous definition for LVI, the difficult detection in routine histological section, the presence of potential confounders, the retrospective nature, and an inadequate sample size in most studies. Therefore, it is necessary to conduct prognostic studies using standardized methods to define and quantify LVI.
Collapse
|
15
|
Tang Q, Efe G, Chiarella AM, Leung J, Chen M, Yamazoe T, Su Z, Pitarresi JR, Li J, Islam M, Karakasheva T, Klein-Szanto AJ, Pan S, Hu J, Natsugoe S, Gu W, Stanger BZ, Wong KK, Diehl JA, Bass AJ, Nakagawa H, Murphy ME, Rustgi AK. Mutant p53 regulates Survivin to foster lung metastasis. Genes Dev 2021; 35:528-541. [PMID: 33737385 PMCID: PMC8015716 DOI: 10.1101/gad.340505.120] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 02/15/2021] [Indexed: 01/01/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most lethal cancers worldwide and evolves often to lung metastasis. P53R175H (homologous to Trp53R172H in mice) is a common hot spot mutation. How metastasis is regulated by p53R175H in ESCC remains to be investigated. To investigate p53R175H-mediated molecular mechanisms, we used a carcinogen-induced approach in Trp53R172H/- mice to model ESCC. In the primary Trp53R172H/- tumor cell lines, we depleted Trp53R172H (shTrp53) and observed a marked reduction in cell invasion in vitro and lung metastasis burden in a tail-vein injection model in comparing isogenic cells (shCtrl). Furthermore, we performed bulk RNA-seq to compare gene expression profiles of metastatic and primary shCtrl and shTrp53 cells. We identified the YAP-BIRC5 axis as a potential mediator of Trp53R172H -mediated metastasis. We demonstrate that expression of Survivin, an antiapoptotic protein encoded by BIRC5, increases in the presence of Trp53R172H Furthermore, depletion of Survivin specifically decreases Trp53R172H-driven lung metastasis. Mechanistically, Trp53R172H but not wild-type Trp53, binds with YAP in ESCC cells, suggesting their cooperation to induce Survivin expression. Furthermore, Survivin high expression level is associated with increased metastasis in several GI cancers. Taken together, this study unravels new insights into how mutant p53 mediates metastasis.
Collapse
Affiliation(s)
- Qiaosi Tang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Gizem Efe
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Anna M Chiarella
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Jessica Leung
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania 19104, USA
| | - Maoting Chen
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Taiji Yamazoe
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Zhenyi Su
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Jason R Pitarresi
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Jinyang Li
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Mirazul Islam
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Tatiana Karakasheva
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Andres J Klein-Szanto
- Department of Pathology, Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19104, USA
| | - Samuel Pan
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Jianhua Hu
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Shoji Natsugoe
- Department of Digestive Surgery, Kagoshima University, Sakuragaoka, Kagoshima 890-0065, Japan
| | - Wei Gu
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Ben Z Stanger
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Kwok-K Wong
- New York University Langone Center, New York, New York 10016, USA
| | - J Alan Diehl
- Case Western University, Cleveland, Ohio 44106, USA
| | - Adam J Bass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Hiroshi Nakagawa
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| | - Maureen E Murphy
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania 19104, USA
| | - Anil K Rustgi
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York 10032, USA
| |
Collapse
|
16
|
Sun Y, Cao S, Li Z, Liu X, Xu J, Tian Y, Shen S, Zhou Y. A novel prognostic factor TIPE2 inhibits cell proliferation and promotes apoptosis in pancreatic ductal adenocarcinoma (PDAC). Int J Med Sci 2021; 18:2051-2062. [PMID: 33850476 PMCID: PMC8040395 DOI: 10.7150/ijms.51497] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 03/01/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Tumor necrosis factor (TNF)-alpha-induced protein 8-like 2 (TIPE2 or TNFAIP8L2) is a newly discovered negative immune regulator. Studies have shown that TIPE2 causes significant malignant biological effects and is differentially expressed in various malignant tumors. However, the expression and roles of TIPE2 in pancreatic ductal adenocarcinoma (PDAC) are largely unknown. Materials and Methods: The expression of TIPE2 in PDAC tissues was assessed by immunohistochemistry, qPCR and western blot analysis and related clinicopathological parameters including survival time were analyzed. After overexpression of TIPE2, cell proliferation and apoptosis analysis were conducted, and the associated underlying molecular mechanism was also explored. Results: In the present study, TIPE2 was upregulated in early PDAC tissues, and TIPE2 expression decreased as the tumor progressed (P<0.001). TIPE2 expression was negatively associated with tumor size, TNM stage and metastasis of lymph nodes. Furthermore, as an independent risk factor, TIPE2 could be used to predict the survival of patients with PDAC (P=0.035). TIPE2 overexpression significantly suppressed the viability, proliferation and induced apoptosis of PDAC cells by inhibiting survivin and increasing the activity of caspase3/7. Conclusions: For the first time, this study demonstrated that TIPE2 is an independent prognostic factor in PDAC. TIPE2 inhibited the proliferation and induced apoptosis via regulating survivin/caspase3/7 signaling pathway. These results indicated that TIPE2 is a potential biomarker for predicting the prognosis of PDAC patients and plays a pivotal role in the progression of PDAC.
Collapse
Affiliation(s)
- Yuqi Sun
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Shougen Cao
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Zequn Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Xiaodong Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Jinxiang Xu
- Department of Hepatology, The First People's Hospital of Luoyang City, Luoyang, Henan, People's Republic of China
| | - Yulong Tian
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Shuai Shen
- Department of Anorectal Surgery, Weifang People's Hospital, Weifang, Shandong, People's Republic of China
| | - Yanbing Zhou
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| |
Collapse
|
17
|
Miao X, Deng Z, Wang S, Weng H, Zhang X, Li H, Xie H, Zhang J, Zhong Y, Zhang B, Li Q, Xie M. IAP-1 promoted cisplatin resistance in nasopharyngeal carcinoma via inhibition of caspase-3-mediated apoptosis. Am J Cancer Res 2021; 11:640-667. [PMID: 33791146 PMCID: PMC7994165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/09/2020] [Indexed: 06/12/2023] Open
Abstract
Recurrent/metastatic nasopharyngeal carcinoma (NPC) is known for having a poor prognosis due to its unfavorable response to chemoradiotherapy. However, the specific processes involved remain poorly understood. This study focused on the cisplatin-resistance mechanism in NPC to help understand the occurrence of advanced NPC and aims to explore the potential therapeutic target for cisplatin-resistant NPC. Two cisplatin-resistant NPC cell lines, HNE-1/DDP and CNE-2/DDP, were established and the differentially expressed genes (DEGs) between parental and cisplatin-resistance cell lines, filtering from high-throughput sequencing results, were analyzed. Next, the effects of IAP-1 on cisplatin-resistant nasopharyngeal cancer cell proliferation, apoptosis, drug resistance and associated cell signaling were evaluated in vitro and in vitro. From our bioinformatic results, more than 15,000 differentially expressed genes (DEGs) were found between parental and resistant cell lines. Nine related DEGs were found in the classic platinum resistance pathway, three of which (ATM, IAP-1, and IAP-2) also appeared in the top five differentially expressed pathways, with elevated IAP-1 showing the highest fold change. Further studies revealed that high IAP-1 expression can lead to an increased cisplatin inhibitory concentration and apoptosis inhibition. IAP-1 silencing can induce upregulation of the caspase-3 and enhance the antiproliferation and proapoptotic effects of cisplatin. Clinical data also showed that IAP-1 overexpression was associated with a worse survival status. In summary, in vitro and in vivo experiments demonstrated that IAP-1 plays a vital role in cisplatin resistance by regulating caspase induced apoptosis and serve as a potential novel therapeutic target and a prognostic indicator for advanced NPC.
Collapse
Affiliation(s)
- Xiangwan Miao
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
| | - Zeyi Deng
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
| | - Siqi Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
| | - Huanhuan Weng
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
| | - Xinting Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
| | - Hailiang Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhuhai People’s HospitalZhuhai 519000, China
| | - Huifen Xie
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
| | - Juan Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
| | - Ying Zhong
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhuhai People’s HospitalZhuhai 519000, China
| | - Bohui Zhang
- Department of Pathology, Zhuhai People’s HospitalZhuhai 519000, China
| | - Quanming Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhuhai People’s HospitalZhuhai 519000, China
| | - Minqiang Xie
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical UniversityGuangzhou 510282, China
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhuhai People’s HospitalZhuhai 519000, China
| |
Collapse
|
18
|
Activation of Serum/Glucocorticoid Regulated Kinase 1/Nuclear Factor-κB Pathway Are Correlated with Low Sensitivity to Bortezomib and Ixazomib in Resistant Multiple Myeloma Cells. Biomedicines 2021; 9:biomedicines9010033. [PMID: 33406639 PMCID: PMC7823718 DOI: 10.3390/biomedicines9010033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/14/2020] [Accepted: 12/30/2020] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is an incurable malignancy often associated with primary and acquired resistance to therapeutic agents, such as proteasome inhibitors. However, the mechanisms underlying the proteasome inhibitor resistance are poorly understood. Here, we elucidate the mechanism of primary resistance to bortezomib and ixazomib in the MM cell lines, KMS-20, KMS-26, and KMS-28BM. We find that low bortezomib and ixazomib concentrations induce cell death in KMS-26 and KMS-28BM cells. However, high bortezomib and ixazomib concentrations induce cell death only in KMS-20 cells. During Gene Expression Omnibus analysis, KMS-20 cells exhibit high levels of expression of various genes, including anti-phospho-fibroblast growth factor receptor 1 (FGFR1), chemokine receptor type (CCR2), and serum and glucocorticoid regulated kinase (SGK)1. The SGK1 inhibitor enhances the cytotoxic effects of bortezomib and ixazomib; however, FGFR1 and CCR2 inhibitors do not show such effect in KMS-20 cells. Moreover, SGK1 activation induces the phosphorylation of NF-κB p65, and an NF-κB inhibitor enhances the sensitivity of KMS-20 cells to bortezomib and ixazomib. Additionally, high levels of expression of SGK1 and NF-κB p65 is associated with a low sensitivity to bortezomib and a poor prognosis in MM patients. These results indicate that the activation of the SGK1/NF-κB pathway correlates with a low sensitivity to bortezomib and ixazomib, and a combination of bortezomib and ixazomib with an SGK1 or NF-κB inhibitor may be involved in the treatment of MM via activation of the SGK1/NF-κB pathway.
Collapse
|
19
|
Santonocito S, Polizzi A, De Pasquale R, Ronsivalle V, Lo Giudice A, Isola G. Analysis of the Efficacy of Two Treatment Protocols for Patients with Symptomatic Oral Lichen Planus: A Randomized Clinical Trial. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 18:E56. [PMID: 33374791 PMCID: PMC7794703 DOI: 10.3390/ijerph18010056] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023]
Abstract
Oral lichen planus (OLP) is a chronic, inflammatory, immune-mediated disease, which can alter the quality of life of patients. The aim of this randomized clinical trial was to compare the therapeutic efficacy of clobetasol oral gel 0.05% versus an anti-inflammatory in oral solution (mouthwash) in the management of patients suffering from symptomatic OLP. The secondary objective was to analyze which one of the two treatments induced a greater risk of developing side effects. Forty patients were assigned (20 patients for group), through a randomized design, to receive clobetasol gel 0.05% or an anti-inflammatory mouthwash, which contains calcium hydroxide, hyaluronic acid, umbelliferone and oligomeric pro-anthocyanidins) for three months. At baseline (T0) and after 3 months (T1), patients underwent dental and dermatological examinations to assess their symptoms (Numerical Pain Scale (NRS) score) and signs (Thongprasom score). Data were calculated using T-test for the dependent variable, Wilcoxon test and Mann-Whitney u test. Both clobetasol and anti-inflammatory resulted in a statistically significant reduction of signs, (p < 0.001 and p = 0.02, respectively) and symptoms (p < 0.001 for clobetasol and p = 0.02 for anti-inflammatory). In conclusion, the results evidenced that, compared to clobetasol, the anti-inflammatory was less effective in determining the reduction of signs and symptom in OLP patients.
Collapse
Affiliation(s)
- Simona Santonocito
- Department of General Surgery and Surgical-Medical Specialties, Unit of Oral Pathology, School of Dentistry, University of Catania, 95124 Catania, Italy; (S.S.); (A.P.); (V.R.); (A.L.G.)
| | - Alessandro Polizzi
- Department of General Surgery and Surgical-Medical Specialties, Unit of Oral Pathology, School of Dentistry, University of Catania, 95124 Catania, Italy; (S.S.); (A.P.); (V.R.); (A.L.G.)
| | - Rocco De Pasquale
- Department of General Surgery and Surgical-Medical Specialties, Unit of Dermatology, University of Catania, 95124 Catania, Italy;
| | - Vincenzo Ronsivalle
- Department of General Surgery and Surgical-Medical Specialties, Unit of Oral Pathology, School of Dentistry, University of Catania, 95124 Catania, Italy; (S.S.); (A.P.); (V.R.); (A.L.G.)
| | - Antonino Lo Giudice
- Department of General Surgery and Surgical-Medical Specialties, Unit of Oral Pathology, School of Dentistry, University of Catania, 95124 Catania, Italy; (S.S.); (A.P.); (V.R.); (A.L.G.)
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialties, Unit of Oral Pathology, School of Dentistry, University of Catania, 95124 Catania, Italy; (S.S.); (A.P.); (V.R.); (A.L.G.)
| |
Collapse
|
20
|
de Campos PS, Menti LD, Pazutti L, Bortoli NÂ, Ferreira LA, van Wyk JL, Darkwa J, Schrekker HS, Lamers ML. The anti-tumor effects of imidazolium salts on oral squamous cell carcinoma. J Oral Pathol Med 2020; 50:470-477. [PMID: 33340378 DOI: 10.1111/jop.13146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/21/2020] [Accepted: 10/24/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Imidazolium salts (IS), ionic derivatives of neutral imidazoles, have properties that can be adjusted by structural modifications to their cations and anions, which makes this particular class of compounds a promising option for developing biologically active compounds. The anti-tumor effects of the IS 1-n-butyl-3-methylimidazolium chloride (C4 MImCl), 1-n-decyl-3-methylimidazolium chloride (C10 MImCl), 1-n-hexadecyl-3-methylimidazolium chloride (C16 MImCl), 1-n-hexadecyl-2,3-dimethylimidazolium chloride (C16 M2 ImCl), 1-n-octadecyl-3-methylimidazolium chloride (C18 MImCl), 1-n-hexadecyl-3-methylimidazolium methanesulfonate (C16 MImMeS), and 1-n-hexadecyl-2,3- dimethylimidazolium methanesulfonate (C16 M2 ImMeS) on oral squamous cell carcinoma (OSCC) have been studied here. METHODS Oral squamous cell carcinoma cells (CAL27) were incubated with increasing IS doses and then submitted to proliferation (2D), cell death (2D) and spheroid assay (3D). RESULTS The IS anti-tumor effect was dependent on both its N-alkyl chain length and anion, whereby C16 MImCl proved to be more effective in combination for inhibiting cell proliferation and cell-cell adhesion, outperforming the methylated C16 M2 ImCl derivative and, most importantly, the gold standard-cisplatin. In addition, C16 MImCl had little effect on keratinocytes and more pronounced effects on more aggressive tumor cells. It also exhibited similar effects on inducing cell death when compared to Cisplatin. This compound spread to a greater area of the tumor sphere and produced an enhanced number of apoptotic and necrotic cells in the tumor cell line, demonstrating only a small rise in the healthy cells. CONCLUSION These data indicate that the effect of C16 MlmCl on OSCC is promising, as it is selective for cancer cells.
Collapse
Affiliation(s)
- Paloma Santos de Campos
- Dentistry School, Basic Research Center, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luiza Deitos Menti
- Dentistry School, Basic Research Center, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luise Pazutti
- Dentistry School, Basic Research Center, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Natália Ângela Bortoli
- Dentistry School, Basic Research Center, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Leonildo Alves Ferreira
- Laboratory of Technological Processes and Catalysis, Institute of Chemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Juanita Lizele van Wyk
- Laboratory of Technological Processes and Catalysis, Institute of Chemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,School of Chemistry, Molecular Science Institute, University of Witswatersrand, Johannesburg, South Africa
| | - James Darkwa
- Department of Chemical Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Henri Stephan Schrekker
- Laboratory of Technological Processes and Catalysis, Institute of Chemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marcelo Lazzaron Lamers
- Dentistry School, Basic Research Center, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Morphological Sciences, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
21
|
Development of a carrier system containing hyaluronic acid and protamine for siRNA delivery in the treatment of melanoma. Invest New Drugs 2020; 39:66-76. [PMID: 32794135 DOI: 10.1007/s10637-020-00986-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/10/2020] [Indexed: 10/23/2022]
Abstract
The use of small interfering RNA (siRNA) in melanoma treatment remains limited owing to its biological properties. Herein, we developed a carrier system containing hyaluronic acid and protamine for siRNA delivery. Considering zeta potential and particle size as standards, the ratio of each component in liposome nanoparticles prepared was screened using the control variable method, and siRNA cationic liposome nanoparticles were prepared based on the optimal results obtained. The encapsulation rate of the cationic liposome nanoparticles was measured, and particle morphology was observed. B16F10 cells were treated with the nanoparticles; 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay, cell scratch experiments, and cell uptake experiments were performed to determine the effectiveness of the loaded siRNA. A mouse model was then established, and tumour tissues were subjected to haematoxylin-eosin staining. The inhibition of the survivin gene and protein expression were assessed using reverse transcription-polymerase chain reaction and western blotting, respectively. The results showed that the optimal mass ratio of hyaluronic acid (HA)-siRNA-to-protamine was 1.0; in the HA-siRNA-protamine complex containing 25 μg siRNA, the addition of 50 μL liposomes yielded optimal particles. And encapsulation rate was 85.07%. The nanoparticles demonstrated a significant inhibitory effect against melanoma cells; siRNA liposomes may inhibit tumour growth by down-regulating survivin. Survivin-siRNA cationic liposome nanoparticles could effectively inhibit the proliferation and migration of melanoma B16F10 cells in vitro and the proliferation of subcutaneous melanoma B16F10 cells, probably by inhibiting survivin mRNA and protein expression. Graphical abstract.
Collapse
|
22
|
Yang CY, Liu CR, Chang IYF, OuYang CN, Hsieh CH, Huang YL, Wang CI, Jan FW, Wang WL, Tsai TL, Liu H, Tseng CP, Chang YS, Wu CC, Chang KP. Cotargeting CHK1 and PI3K Synergistically Suppresses Tumor Growth of Oral Cavity Squamous Cell Carcinoma in Patient-Derived Xenografts. Cancers (Basel) 2020; 12:cancers12071726. [PMID: 32610557 PMCID: PMC7408003 DOI: 10.3390/cancers12071726] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/23/2020] [Accepted: 06/26/2020] [Indexed: 01/27/2023] Open
Abstract
Oral cavity squamous cell carcinomas (OSCCs) are aggressive tumors, and their recurrence leads to poor prognosis and reduced survival rates. This study aimed to identify therapeutic targets and to evaluate the efficacy of targeted inhibitors in OSCC patient-derived xenograft (PDX) models. Herein, we reported that OSCC PDXs recapitulated the genomic signatures of their paired primary tumors and the expression of CHEK1, PIK3CA, and PIK3CD was significantly upregulated in OSCC. The antitumor efficacy of CHK1 inhibitors (PF477736, AZD7762, LY2606368) and PI3K inhibitors (BYL719, GDC0941, GSK1059615) was investigated in OSCC cell lines and PDX models. Targeting either CHK1 or PI3K effectively inhibited cell proliferation and colony formation by inducing cell cycle arrest and apoptosis in in vitro cell-based assays. Cisplatin-based chemotherapy combined with CHK1 inhibitor treatment synergistically inhibited cell proliferation by suppressing CHK1 phosphorylation and inducing PARP cleavage. Furthermore, compared with monotherapy, cotreatment with CHK1 and PI3K inhibitors exerted synergistic anticancer effects by suppressing CHK1, AKT, and 4E-BP1 phosphorylation. In summary, our study identified CHK1 and PI3K as promising targets, especially in a dual treatment strategy combining a CHK1 inhibitor with cisplatin or a PI3K inhibitor as a novel therapeutic approach for OSCC patients with aberrant cell cycle regulation and PI3K signaling activation.
Collapse
Affiliation(s)
- Chia-Yu Yang
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-Y.Y.); (C.-R.L.); (F.-W.J.); (W.-L.W.); (T.-L.T.)
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (H.L.); (Y.-S.C.)
- Department of Otolaryngology Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 33302, Taiwan; (I.Y.-F.C.); (C.-N.O.)
| | - Chiao-Rou Liu
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-Y.Y.); (C.-R.L.); (F.-W.J.); (W.-L.W.); (T.-L.T.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Ian Yi-Feng Chang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 33302, Taiwan; (I.Y.-F.C.); (C.-N.O.)
| | - Chun-Nan OuYang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 33302, Taiwan; (I.Y.-F.C.); (C.-N.O.)
| | - Chia-Hsun Hsieh
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yen-Lin Huang
- Department of Pathology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Chun-I Wang
- Department of Otolaryngology Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Fei-Wen Jan
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-Y.Y.); (C.-R.L.); (F.-W.J.); (W.-L.W.); (T.-L.T.)
| | - Wan-Ling Wang
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-Y.Y.); (C.-R.L.); (F.-W.J.); (W.-L.W.); (T.-L.T.)
| | - Ting-Lin Tsai
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-Y.Y.); (C.-R.L.); (F.-W.J.); (W.-L.W.); (T.-L.T.)
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (H.L.); (Y.-S.C.)
| | - Hsuan Liu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (H.L.); (Y.-S.C.)
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 33302, Taiwan; (I.Y.-F.C.); (C.-N.O.)
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Ching-Ping Tseng
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yu-Sun Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (H.L.); (Y.-S.C.)
- Department of Otolaryngology Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 33302, Taiwan; (I.Y.-F.C.); (C.-N.O.)
| | - Chih-Ching Wu
- Department of Otolaryngology Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 33302, Taiwan; (I.Y.-F.C.); (C.-N.O.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Correspondence: (C.-C.W.); or (K.-P.C.)
| | - Kai-Ping Chang
- Department of Otolaryngology Head and Neck Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 33302, Taiwan; (I.Y.-F.C.); (C.-N.O.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Correspondence: (C.-C.W.); or (K.-P.C.)
| |
Collapse
|
23
|
Circumventing AKT-Associated Radioresistance in Oral Cancer by Novel Nanoparticle-Encapsulated Capivasertib. Cells 2020; 9:cells9030533. [PMID: 32106632 PMCID: PMC7140405 DOI: 10.3390/cells9030533] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/11/2020] [Accepted: 02/24/2020] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Development of radioresistance in oral squamous cell carcinoma (OSCC) remains a significant problem in cancer treatment, contributing to the lack of improvement in survival trends in recent decades. Effective strategies to overcome radioresistance are necessary to improve the therapeutic outcomes of radiotherapy in OSCC patients. METHODS Cells and xenograft tumors were irradiated using the Small Animal Radiation Research Platform. AKT inhibitor capivasertib (AZD5363) was encapsulated into cathepsin B-responsible nanoparticles (NPs) for tumor-specific delivery. Cell viability was measured by alamarBlue, cell growth was determined by colony formation and 3D culture, and apoptosis was assessed by flow cytometry with the staining of Fluorescein isothiocyanate (FITC) Annexin V and PI. An orthotopic tongue tumor model was used to evaluate the in vivo therapeutic effects. The molecular changes induced by the treatments were assessed by Western blotting and immunohistochemistry. RESULTS We show that upregulation of AKT signaling is the critical mechanism for radioresistance in OSCC cells, and AKT inactivation by a selective and potent AKT inhibitor capivasertib results in radiosensitivity. Moreover, relative to irradiation (IR) alone, IR combined with the delivery of capivasertib in association with tumor-seeking NPs greatly enhanced tumor cell repression in 3D cell cultures and OSCC tumor shrinkage in an orthotopic mouse model. CONCLUSIONS These data indicate that capivasertib is a potent agent that sensitizes radioresistant OSCC cells to IR and is a promising strategy to overcome failure of radiotherapy in OSCC patients.
Collapse
|
24
|
Efficient induction of cytotoxic T lymphocytes in hepatocellular carcinoma using the HLA-A2-restricted survivin peptide in vitro. Exp Cell Res 2020; 386:111741. [PMID: 31759968 DOI: 10.1016/j.yexcr.2019.111741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 11/17/2019] [Accepted: 11/19/2019] [Indexed: 11/27/2022]
Abstract
Survivin is a newly identified tumour-associated antigen and has been demonstrated to be an excellent target for immunotherapy in several cancers, but its role in hepatocellular carcinoma treatment is still unknown. In this study, survivin-derived peptide-specific cytotoxic T lymphocytes (CTLs) from peripheral blood mononuclear cells of healthy donors were induced by multiple stimulations with HLA-A2- restricted survivin peptide-pulsed T2 cells. The induced CTLs exhibited specific lysis of T2 cells pulsed with the peptide and HLA-A2+ hepatocellular carcinoma cells expressing survivin, while HLA-A2+ hepatocellular carcinoma cell lines that did not express survivin were not recognized by the CTLs. These results suggest that the survivin peptide epitope could be a potential target of specific immunotherapy for HLA-A2+ patients with hepatocellular carcinoma.
Collapse
|
25
|
Goldie SJ, Chincarini G, Darido C. Targeted Therapy Against the Cell of Origin in Cutaneous Squamous Cell Carcinoma. Int J Mol Sci 2019; 20:ijms20092201. [PMID: 31060263 PMCID: PMC6539622 DOI: 10.3390/ijms20092201] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 04/29/2019] [Accepted: 04/29/2019] [Indexed: 01/03/2023] Open
Abstract
Squamous cell carcinomas (SCC), including cutaneous SCCs, are by far the most frequent cancers in humans, accounting for 80% of all newly diagnosed malignancies worldwide. The old dogma that SCC develops exclusively from stem cells (SC) has now changed to include progenitors, transit-amplifying and differentiated short-lived cells. Accumulation of specific oncogenic mutations is required to induce SCC from each cell population. Whilst as fewer as one genetic hit is sufficient to induce SCC from a SC, multiple events are additionally required in more differentiated cells. Interestingly, the level of differentiation correlates with the number of transforming events required to induce a stem-like phenotype, a long-lived potential and a tumourigenic capacity in a progenitor, a transient amplifying or even in a terminally differentiated cell. Furthermore, it is well described that SCCs originating from different cells of origin differ not only in their squamous differentiation status but also in their malignant characteristics. This review summarises recent findings in cutaneous SCC and highlights transforming oncogenic events in specific cell populations. It underlines oncogenes that are restricted either to stem or differentiated cells, which could provide therapeutic target selectivity against heterogeneous SCC. This strategy may be applicable to SCC from different body locations, such as head and neck SCCs, which are currently still associated with poor survival outcomes.
Collapse
Affiliation(s)
- Stephen J Goldie
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5001, Australia.
| | - Ginevra Chincarini
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
| | - Charbel Darido
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
26
|
Metastatic Cutaneous Squamous Cell Cancer with Peritoneal Carcinomatosis: A Case Report. REPORTS 2019. [DOI: 10.3390/reports2010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Peritoneal involvement as a metastatic site of squamous cell skin cancer is exceptionally rare. The current work analyzes a 52-year old male with high-risk cutaneous squamous cell nose carcinoma (cSCC) that was initially treated with surgery and platinum-based concurrent chemoradiation. Five years later, he presented jaundice and hypercalcemia. Further imaging revealed diffused liver, peritoneal and paraaortic lymph node metastases without evidence of locoregional recurrence. The patient underwent liver biopsy, which confirmed the diagnosis. High-risk features for metastasizing can be considered the maximum clinical diameter, the anatomical subsite (localization of the primary tumor in the ear and retroauricular area, cheek and lip are considered to significantly increase the risk of distant metastasis), poor histological differentiation, perineural invasion and lesions with a thickness of more than 2.0 mm. Late relapse that involves only disseminated abdominal disease is very uncommon and may justify closer follow-up and more aggressive chemotherapy in high-risk patients.
Collapse
|
27
|
Laka K, Makgoo L, Mbita Z. Survivin Splice Variants in Arsenic Trioxide (As₂O₃)-Induced Deactivation of PI3K and MAPK Cell Signalling Pathways in MCF-7 Cells. Genes (Basel) 2019; 10:genes10010041. [PMID: 30646589 PMCID: PMC6356383 DOI: 10.3390/genes10010041] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/30/2018] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
Several pathways are deregulated during carcinogenesis but most notably, tumour cells can lose cell cycle control and acquire resistance to apoptosis by expressing a number of anti-apoptotic proteins such as the Inhibitors of Apoptosis Protein (IAP) family of proteins that include survivin, which is implicated in cancer development. There is no study which had proven that arsenic trioxide (As2O3) has any effect on the splicing machinery of survivin and its splice variants, hence this study was aimed at determining the cytotoxic effect of As2O3 and its effect on the expression pattern of survivin splice variants in MCF-7 cells. As2O3 inhibited the growth of the MCF-7 cells in a concentration-dependent manner. The Muse® Cell Analyser showed that As2O3-induced G2/M cell cycle arrest, promoted caspase-dependent apoptosis without causing any damage to the mitochondrial membrane of MCF-7 cells. As2O3 also deactivated two survival pathways, Mitogen-Activated Protein Kinase (MAPK) and Phosphoinositide 3-Kinase (PI3K) signalling pathways in MCF-7 cells. Deactivation of the two pathways was accompanied by the upregulation of survivin 3α during As2O3-induced G2/M cell cycle arrest and apoptosis. Survivin 2B was found to be upregulated only during As2O3-induced G2/M cell cycle arrest but downregulated during As2O3-induced apoptosis. Survivin wild-type was highly expressed in the untreated MCF-7 cells, the expression was upregulated during As2O3-induced G2/M cell cycle arrest and it was downregulated during As2O3-induced apoptosis. Survivin variant ΔEx3 was undetected in both untreated and treated MCF-7 cells. Survivin proteins were localised in both the nucleus and cytoplasm in MCF-7 cells and highly upregulated during the As2O3-induced G2/M cell cycle arrest, which can be attributed to the upregulation of survivin-2B. This study has provided the first evidence showing that the novel survivin 2B splice variant may be involved in the regulation of As2O3-induced G2/M cell cycle arrest only. This splice variant can therefore, be targeted for therapeutic purposes against Luminal A breast cancer cells.
Collapse
Affiliation(s)
- Kagiso Laka
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X1106, Sovenga 0727, Polokwane, South Africa.
| | - Lilian Makgoo
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X1106, Sovenga 0727, Polokwane, South Africa.
| | - Zukile Mbita
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X1106, Sovenga 0727, Polokwane, South Africa.
| |
Collapse
|
28
|
Eren-Keleş E, Karabulut HG, Çakmaklı HF, Adaklı B, Köse SK, Uğur-Dinçaslan H, Yavuz G, Ertem M, Tükün A. Expression of Survivin and Its Splice Variants in Pediatric Acute Lymphoblastic Leukemia. Genet Test Mol Biomarkers 2018; 22:680-685. [PMID: 30489176 DOI: 10.1089/gtmb.2018.0152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aims: Survivin is involved in the inhibition of apoptosis and the regulation of cell division. In addition to wild-type survivin (survivin-wt), at least four splice variants with differential functions (ΔEx3 and 3B antiapoptotic, and 2α and 2B proapoptotic) have been identified. Survivin is highly expressed in several cancers, including hematological malignancies. Although acute lymphoblastic leukemia (ALL) is the most frequent malignancy in children, studies that investigated survivin expression in ALL are limited, and there is no study on 3B and 2α expression in ALL. Therefore the expression of survivin-wt and its splice variants was investigated in pediatric B-cell ALL patients. Materials and Methods: The expression of survivin-wt and its four splice variants was investigated by quantitative real-time polymerase chain reaction in archival RNA samples of 35 pediatric B-cell ALL patients. Patients were divided into high- and standard-risk groups according to age, white blood cell count, extramedullary involvement, and genetic risk factors; expression of survivin variants was compared between these two risk groups. Results: We found that the ratio of survivin-ΔEx3/wild type (WT) expression was higher in the low-risk group than in the high-risk group. Conclusion: Comparative analysis between the high- and low-risk B-cell ALL groups indicated that the survivin-ΔEx3/WT expression ratio could potentially be used in risk classification for pediatric B-cell ALL.
Collapse
Affiliation(s)
- Efsun Eren-Keleş
- Central Laboratory, Biotechnology Institute, Ankara University, Ankara, Turkey
| | | | - Hasan Fatih Çakmaklı
- Department of Pediatric Hematology and Oncology, School of Medicine, Ankara University, Ankara, Turkey
| | - Başak Adaklı
- Department of Pediatric Hematology and Oncology, Istinye University, Medicalpark Bahçelievler Hospital, Istanbul, Turkey
| | - Serdar Kenan Köse
- Department of Biostatistics, School of Medicine, Ankara University, Ankara, Turkey
| | - Handan Uğur-Dinçaslan
- Department of Pediatric Hematology and Oncology, School of Medicine, Ankara University, Ankara, Turkey
| | - Gülsan Yavuz
- Department of Pediatric Hematology and Oncology, School of Medicine, Ankara University, Ankara, Turkey
| | - Mehmet Ertem
- Department of Pediatric Hematology and Oncology, School of Medicine, Ankara University, Ankara, Turkey
| | - Ajlan Tükün
- Duzen Laboratories Group, Division of Medical Genetics, Ankara, Turkey
| |
Collapse
|
29
|
López-Verdín S, Lavalle-Carrasco J, Carreón-Burciaga RG, Serafín-Higuera N, Molina-Frechero N, González-González R, Bologna-Molina R. Molecular Markers of Anticancer Drug Resistance in Head and Neck Squamous Cell Carcinoma: A Literature Review. Cancers (Basel) 2018; 10:cancers10100376. [PMID: 30308958 PMCID: PMC6210289 DOI: 10.3390/cancers10100376] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/01/2018] [Accepted: 10/08/2018] [Indexed: 02/07/2023] Open
Abstract
This manuscript provides an update to the literature on molecules with roles in tumor resistance therapy in head and neck squamous cell carcinoma (HNSCC). Although significant improvements have been made in the treatment for head and neck squamous cell carcinoma, physicians face yet another challenge-that of preserving oral functions, which involves the use of multidisciplinary therapies, such as multiple chemotherapies (CT) and radiotherapy (RT). Designing personalized therapeutic options requires the study of genes involved in drug resistance. This review provides an overview of the molecules that have been linked to resistance to chemotherapy in HNSCC, including the family of ATP-binding cassette transporters (ABCs), nucleotide excision repair/base excision repair (NER/BER) enzymatic complexes (which act on nonspecific DNA lesions generated by gamma and ultraviolet radiation by cross-linking and forming intra/interchain chemical adducts), cisplatin (a chemotherapeutic agent that causes DNA damage and induces apoptosis, which is a paradox because its effectiveness is based on the integrity of the genes involved in apoptotic signaling pathways), and cetuximab, including a discussion of the genes involved in the cell cycle and the proliferation of possible markers that confer resistance to cetuximab.
Collapse
Affiliation(s)
- Sandra López-Verdín
- Research Institute of Dentistry, Health Science Center, Universidad de Guadalajara, Guadalajara 4430, JAL, Mexico.
| | - Jesús Lavalle-Carrasco
- Department of Research, School of Dentistry, Universidad Juárez del Estado de Durango, Durango 34000, DGO, Mexico.
| | - Ramón G Carreón-Burciaga
- Department of Research, School of Dentistry, Universidad Juárez del Estado de Durango, Durango 34000, DGO, Mexico.
| | - Nicolás Serafín-Higuera
- Molecular Biology Department, School of Dentistry, Universidad Autónoma de Baja California, Mexicali 21040, Mexico.
| | - Nelly Molina-Frechero
- Department of Health Care, Xochimilco Unit, Universidad Autónoma Metropolitana (UAM) Xochimilco, Mexico City 04960, Mexico.
| | - Rogelio González-González
- Department of Research, School of Dentistry, Universidad Juárez del Estado de Durango, Durango 34000, DGO, Mexico.
| | - Ronell Bologna-Molina
- Department of Research, School of Dentistry, Universidad Juárez del Estado de Durango, Durango 34000, DGO, Mexico.
- Molecular Pathology Area, School of Dentistry, Universidad de la República, Montevideo 11600, Uruguay.
| |
Collapse
|
30
|
Zhou L, Lu J, Liang ZY, Zhou WX, Yuan D, Li BQ, You L, Guo JC, Zhao YP. High nuclear Survivin expression as a poor prognostic marker in pancreatic ductal adenocarcinoma. J Surg Oncol 2018; 118:1115-1121. [PMID: 30261114 DOI: 10.1002/jso.25253] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 09/04/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Survivin, one of the key regulators of mitosis and apoptosis, has long been well recognized to play important biological roles in many neoplasms, including pancreatic ductal adenocarcinoma (PDAC). However, its prognostic value in PDAC remains controversial. PATIENTS AND METHODS Nuclear expression of Survivin was detected, using tissue microarray-based immunohistochemistry, in paired-tumor and nontumor samples from 306 patients with radically resected PDAC. The staining H scores were further correlated with clinicopathologic features and disease-specific survival (DSS). RESULTS Nuclear Survivin expression was much higher in tumor than in nontumor tissues (P < 0.001). No significant association between tumoral Survivin expression and clinicopathologic variables was found. For prognosis, high Survivin expression was associated with shortened DSS in all eligible patients and four subgroups, that is, male and nondiabetic patients as well as those with head-located and G1-2 tumors, shown by univariate analyses. In addition, a statistically marginal significance was revealed in eight subgroups. For the entire cohort and two subgroups, nuclear Survivin expression was also multivariate identified as an independent predictor for DSS. For patients with G1-2 tumors, it was the single prognostic marker. CONCLUSION Our data suggest an association between high nuclear Survivin expression and poor prognosis in PDAC. However, further confirmation might be necessary.
Collapse
Affiliation(s)
- Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Jun Lu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Zhi-Yong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Wei-Xun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Da Yuan
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Bing-Qi Li
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Jun-Chao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Yu-Pei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| |
Collapse
|
31
|
Ma W, Xie X, Shao X, Zhang Y, Mao C, Zhan Y, Zhao D, Liu M, Li Q, Lin Y. Tetrahedral DNA nanostructures facilitate neural stem cell migration via activating RHOA/ROCK2 signalling pathway. Cell Prolif 2018; 51:e12503. [PMID: 30091500 DOI: 10.1111/cpr.12503] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 06/20/2018] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES The main purpose of current study was to explore the effects of tetrahedral DNA nanostructures (TDNs) on neuroectodermal (NE-4C) stem cells migration and unveil the potential mechanisms. MATERIALS AND METHODS The successfully self-assembled TDNs were also determined by dynamic light scattering (DLS). A bidirectional wound-healing assay and transwell chamber assay were employed to test the migrating behaviour of NE-4C stem cells cultured under different conditions. RESULTS Through an in vitro study, we found that stem cells could internalize TDNs quickly, and the cells' parallel and vertical migration was promoted effectively. Besides, the effects of TDNs were found being exerted by upregulating the gene and protein expression levels of RhoA, Rock2 and Vinculin, indicating that the RHOA/ROCK2 pathway was activated by the TDNs during the cell migration. CONCLUSIONS In conclusion, TDNs could enter NSCs without the aid of other transfection reagents in large amounts, whereas only small amounts of ssDNA could enter the cells. TDNs taken up by NSCs activated the RHOA/ROCK2 signalling pathway, which had effects on the relevant genes and proteins expression, eventually promoting the migration of NE-4C stem cells. These findings suggested that TDNs have great potential in application for the repair and regeneration of neural tissue.
Collapse
Affiliation(s)
- Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xueping Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoru Shao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuxin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenchen Mao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuxi Zhan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dan Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mengting Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianshun Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
32
|
Aspirin is Involved in the Cell Cycle Arrest, Apoptosis, Cell Migration, and Invasion of Oral Squamous Cell Carcinoma. Int J Mol Sci 2018; 19:ijms19072029. [PMID: 30002310 PMCID: PMC6073368 DOI: 10.3390/ijms19072029] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/05/2018] [Accepted: 07/07/2018] [Indexed: 01/04/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common cancers worldwide. In China, its 5-year survival rate is roughly 50%, owing to acquired chemotherapeutic resistance and metastasis of the disease. Accumulating evidence demonstrates that aspirin (ASA) acts as a preventive or therapeutic agent in multiple cancers; however, anti-tumor activities induced by aspirin are unclear in OSCC. To investigate the possible role of aspirin in OSCC development, we first employed bioinformatics to analyze the anti-OSCC effects of aspirin. We performed a genetic oncology (GO) enrichment analysis using the Database for Annotation, Visualization, and Integrated Discovery (DAVID), and the protein⁻protein interaction (PPI) network analysis by Cytoscape for differentially expressed genes (DEGs). We also evaluated the potential effects of aspirin on cell proliferation, invasion, migration, and apoptosis in two well-characterized OSCC cell lines (TCA8113 and CAL27). The bioinformatic results revealed that aspirin could inhibit proliferation by blocking the cell cycle, and could reduce migration and invasion via the PI3K-Akt and focal adhesion pathways. We found that ASA could downregulate the OSCC cell proliferation colony formation, invasion, and migration, as well as upregulate apoptosis. Furthermore, we found that ASA suppressed the activation of the focal adhesion kinase (FAK) and the phosphorylation of Akt, NF-κB, and STAT3. Overall, our data suggested that ASA may be developed as a chemopreventive agent to effectively treat OSCC.
Collapse
|