1
|
Apaydın E, Yaşar B, Şimşek G, Kaygın P, Sarıaltın SY, Dirican O, Çetin HE, Husseini AA, Oğuztüzün S. The Administration of Steroids and its Impact on Caspase-3 Expression in Pediatric Adenoid Hypertrophy. Indian J Otolaryngol Head Neck Surg 2024; 76:4516-4522. [PMID: 39376326 PMCID: PMC11456078 DOI: 10.1007/s12070-024-04900-8] [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: 12/11/2023] [Accepted: 07/07/2024] [Indexed: 10/09/2024] Open
Abstract
Objective Adenoid hypertrophy is a prevalent pediatric condition, often necessitating surgical intervention. Intranasal steroid administration shows promise as a conservative treatment, particularly by inducing apoptosis in adenoidal cells, leading to a reduction in adenoid size and inflammation. This study aims to characterize the expression profile of caspase-3 as an apoptotic inducer protein in inflammatory and epithelial adenoid tissues and explore its association with steroid administration. Methods We performed immunohistochemical staining for caspase-3 proteins in adenoid tissues obtained from 51 pediatric patients aged between 2.5 and 12 years (mean age: 6.09 ± 2.1 years) who underwent adenoid surgery. A retrospective analysis of clinical data was conducted, categorizing participants into steroid treatment receivers (n = 25) and non-receivers (n = 26). Subsequently, the lymphoid inflammatory tissue and epithelial tissue from the adenoid were compared in terms of caspase-3 protein expression, and associated clinical variables were assessed. Results Immunohistochemical analysis revealed significant caspase-3 expression in inflammatory tissues. The expression levels were scored, and no significant correlation was observed between inflammation and epithelium based on caspase-3 expression (correlation coefficient = 0.143; p > 0.05). Furthermore, demographic and clinical characteristics did not show a statistically significant difference in caspase-3 expression levels. Conclusion Caspase-3 expression was significant in inflammatory adenoid tissue, but it showed no association with nasal steroid administration.
Collapse
Affiliation(s)
- Emre Apaydın
- Department of Otorhinolaryngology, Health Sciences University, Atatürk Sanatorium Training and Research Hospital, Ankara, Türkiye
| | - Buse Yaşar
- Department of Otorhinolaryngology, Health Sciences University, Atatürk Sanatorium Training and Research Hospital, Ankara, Türkiye
| | - Gülçin Şimşek
- Departmet of Pathology, University of Health Sciences, Gulhane Training and Research Hospital, Ankara, Türkiye
| | - Pınar Kaygın
- Faculty of Engineering and Natural Sciences, Department of Biology, Kırıkkale University, Kırıkkale, Türkiye
| | - Sezen Yılmaz Sarıaltın
- Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Ankara University, Ankara, Türkiye
| | - Onur Dirican
- Department of Pathology Laboratory Techniques, Vocational School of Health Services, Istanbul Gelişim University, İstanbul, Türkiye
| | - Hazal Eylem Çetin
- Faculty of Engineering and Natural Sciences, Department of Biology, Kırıkkale University, Kırıkkale, Türkiye
| | - Abbas Ali Husseini
- Life Science and Biomedical Engineering Application and Research Center, Istanbul Gelişim University, İstanbul, Türkiye
| | - Serpil Oğuztüzün
- Faculty of Engineering and Natural Sciences, Department of Biology, Kırıkkale University, Kırıkkale, Türkiye
| |
Collapse
|
2
|
Tajbakhsh A, Yousefi F, Farahani N, Savardashtaki A, Reiner Ž, Jamialahmadi T, Sahebkar A. Molecular Mechanisms and Therapeutic Potential of Resolvins in Cancer - Current Status and Perspectives. Curr Med Chem 2024; 31:5898-5917. [PMID: 37497711 DOI: 10.2174/0929867331666230727100123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 05/26/2023] [Accepted: 06/15/2023] [Indexed: 07/28/2023]
Abstract
Resolvins are specialized pro-resolving mediators derived from omega-3 fatty acids that can suppress several cancer-related molecular pathways, including important activation of transcription parameters in the tumor cells and their microenvironment, inflammatory cell infiltration, cytokines as well as chemokines. Recently, an association between resolvins and an important anti-inflammatory process in apoptotic tumor cell clearance (efferocytosis) was shown. The inflammation status or the oncogene activation increases the risk of cancer development via triggering the transcriptional agents, including nuclear factor kappa-light-chain-enhancer of activated B cells by generating the pro-inflammatory lipid molecules and infiltrating the tumor cells along with the high level of pro-inflammatory signaling. These events can cause an inflammatory microenvironment. Resolvins might decrease the leukocyte influx into the inflamed tissues. It is widely accepted that resolvins prohibit the development of debris-triggered cancer via increasing the clearance of debris, especially by macrophage phagocytosis in tumors without any side effects. Resolvins D2, D1, and E1 might suppress tumor-growing inflammation by activation of macrophages clearance of cell debris in the tumor. Resolvin D5 can assist patients with pain during treatment. However, the effects of resolvins as anti-inflammatory mediators in cancers are not completely explained. Thus, based on the most recent studies, we tried to summarize the most recent knowledge on resolvins in cancers.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Yousefi
- Department of Biological Sciences, Faculty of Genetics, Tarbiat Modares University, Tehran, Iran
| | - Najmeh Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Savardashtaki
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Željko Reiner
- Department of Internal Medicine, University Hospital Center Zagreb, School of Medicine, University of Zagreb, Zagreb, Croatia
- Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Sheinin M, Mondal S, Pahan K. Neutralization of p40 Homodimer and p40 Monomer Leads to Tumor Regression in Patient-Derived Xenograft Mice with Pancreatic Cancer. Cancers (Basel) 2023; 15:5796. [PMID: 38136341 PMCID: PMC10742282 DOI: 10.3390/cancers15245796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/23/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Pancreatic cancer is a highly aggressive cancer with a high mortality rate and limited treatment options. It is the fourth leading cause of cancer in the US, and mortality is rising rapidly, with a 12% relative 5-year survival rate. Early diagnosis remains a challenge due to vague symptoms, lack of specific biomarkers, and rapid tumor progression. Interleukin-12 (IL-12) is a central cytokine that regulates innate (natural killer cells) and adaptive (cytokine T-lymphocytes) immunity in cancer. We demonstrated that serum levels of IL-12p40 homodimer (p402) and p40 monomer (p40) were elevated and that of IL-12 and IL-23 were lowered in pancreatic cancer patients compared to healthy controls. Comparably, human PDAC cells produced greater levels of p402 and p40 and lower levels of IL-12 and IL-23 compared to normal pancreatic cells. Notably, neutralization of p402 by mAb a3-1d and p40 by mAb a3-3a induced the death of human PDAC cells, but not normal human pancreatic cells. Furthermore, we demonstrated that treatment of PDX mice with p402 mAb and p40 mAb resulted in apoptosis and tumor shrinkage. This study illustrates a new role of p402 and p40 monomer in pancreatic cancer, highlighting possible approaches against this deadly form of cancer with p402 and p40 monomer immunotherapies.
Collapse
Affiliation(s)
- Monica Sheinin
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA (S.M.)
| | - Susanta Mondal
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA (S.M.)
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA (S.M.)
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
4
|
Chung AK, Lin RT, Yeh CC, Yang CY, Wu CJ, Chen PL, Lin JT. Diagnostic rate of germline pathogenic variants in pancreatic ductal adenocarcinoma patients using whole genome sequencing. Front Genet 2023; 14:1172365. [PMID: 37234870 PMCID: PMC10205989 DOI: 10.3389/fgene.2023.1172365] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Identification of germline pathogenic variants in cancer patients is critical for treatment planning, genetic counseling, and health policymaking. However, previous estimates of the prevalence of germline etiology of pancreatic ductal adenocarcinoma (PDAC) were biased because they were based only on sequencing data of protein-coding regions of known PDAC candidate genes. To determine the percentage of patients with PDAC carrying germline pathogenic variants, we enrolled the inpatients from the digestive health clinics, hematology and oncology clinics, and surgical clinics of a single tertiary medical center in Taiwan for whole genome sequencing (WGS) analysis of genomic DNA. The virtual gene panel of 750 genes comprised PDAC candidate genes and those listed in the COSMIC Cancer Gene Census. The genetic variant types under investigation included single nucleotide substitutions, small indels, structural variants, and mobile element insertions (MEIs). In 8 of 24 (33.3%) patients with PDAC, we identified pathogenic/likely pathogenic variants, including single nucleotide substitutions and small indels in ATM, BRCA1, BRCA2, POLQ, SPINK1 and CASP8, as well as structural variants in CDC25C and USP44. We identified additional patients carrying variants that could potentially affect splicing. This cohort study demonstrates that an extensive analysis of the abundant information yielded by the WGS approach can uncover many pathogenic variants that could be missed by traditional panel-based or whole exome sequencing-based approaches. The percentage of patients with PDAC carrying germline variants might be much higher than previously expected.
Collapse
Affiliation(s)
- An-Ko Chung
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ro-Ting Lin
- Department of Occupational Safety and Health, College of Public Health, China Medical University, Taichung, Taiwan
| | - Chun-Chieh Yeh
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chi-Ying Yang
- Department of Internal Medicine, Digestive Medicine Center, China Medical University Hospital, Taichung, Taiwan
| | - Chang-Jiun Wu
- Department of Genomic Medicine, MD Anderson Cancer Center, University of Texas, Houston, TX, United States
| | - Pei-Lung Chen
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Jaw-Town Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-Da Hospital, Kaohsiung, Taiwan
| |
Collapse
|
5
|
Srinivasan R, Kamalanathan D, Rathinavel T, Iqbal MN, Shanmugam G. Anti-cancer potentials of aervine validated through in silico molecular docking, dynamics simulations, pharmacokinetic prediction and in vitro assessment of caspase – 3 in SW480 cell line. MOLECULAR SIMULATION 2023. [DOI: 10.1080/08927022.2023.2193646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/01/2023]
|
6
|
Polyethyleneglycol-Betulinic Acid (PEG-BA) Polymer-Drug Conjugate Induces Apoptosis and Antioxidation in a Biological Model of Pancreatic Cancer. Polymers (Basel) 2023; 15:polym15020448. [PMID: 36679328 PMCID: PMC9863557 DOI: 10.3390/polym15020448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most aggressive solid malignancies with poor treatment response and low survival rates. Herbal medicines such as betulinic acid (BA) have shown potential in treating various solid tumours, but with limitations that can be circumvented by polymer-drug conjugation. Polyethylene glycol-BA (PEG-BA) polymer-drug conjugate has previously shown selective anticancer activity against PC cells. Here, we elucidate the mechanism of cell death and the cell death pathway, anti-inflammatory and antioxidant activities of PEG-BA. PEG-BA induced apoptotic cell death by arresting MIA-PaCa-2 cells in the Sub-G1 phase of the cell cycle compared with BA and untreated cells (39.50 ± 5.32% > 19.63 ± 4.49% > 4.57 ± 0.82%). NFκB/p65 protein expression was moderately increased by PEG-BA (2.70 vs. 3.09 ± 0.42 ng/mL; p = 0.1521). However, significant (p < 0.05) overexpression of the proapoptotic genes TNF (23.72 ± 1.03) and CASPASE 3 (12,059.98 ± 1.74) compared with untreated cells was notable. The antioxidant potential of PEG-BA was greater (IC50 = 15.59 ± 0.64 µM) compared with ascorbic acid (25.58 ± 0.44 µM) and BA-only (>100 µM) and further confirmed with the improved reduction of hydroperoxide levels compared with BA-only (518.80 ± 25.53 µM vs. 542.43 ± 9.70 µM). In conclusion, PEG-BA activated both the intrinsic and extrinsic pathways of apoptosis and improved antioxidant activities in PC cells, suggesting enhanced anticancer activity upon conjugation.
Collapse
|
7
|
Mohamed Abdelgawad L, Abdullatif Abdelaziz A, Bawdy El-Begawey M, Mohamed Saafan A. Influence of Nanocurcumin and Photodynamic Therapy Using Nanocurcumin in Treatment of Rat Tongue Oral Squamous Cell Carcinoma Through Histological Examination and Gene Expression of BCL2 and Caspase-3. Rep Biochem Mol Biol 2023; 11:730-738. [PMID: 37131902 PMCID: PMC10149134 DOI: 10.52547/rbmb.11.4.730] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/30/2022] [Indexed: 05/04/2023]
Abstract
Background Oral squamous cell carcinoma (OSCC) is the sixth most common mouth cancer in the world. The aim of the present study is comparing the effects of using Nanocurcumin, and photodynamic therapy (PDT), alone or together in treatment of OSCC in rats. Methods Forty Wister male rats were divided into Control (group 1), 650 nm diode Laser only (group 2), Nanocurcumin alone (group 3), and PDT with a combination of laser with Nanocurcumin (group 4). Then, OSCC in the tongue induced by dimethylbenz anthracene (DMBA). The treatments were evaluated clinically, histopathologically, and immunohistochemically through BCL2 and Caspase-3 genes expression. Results Positive control with OSCC displayed significant weight loss, while PDT group gained more than nanocurcumin treated groups as well as laser groups comparing with control positive group. The histological examination of the tongue in PDT group showed improvement. In laser group, there were partial loss of surface epithelium with various ulcers and dysplasia and partial improvement by this type of treatment. The tongue in the positive control group showed ulcer in the dorsum surface with inflammatory cells, hyperplasia of the mucosa membrane around the ulcer (acanthosis) with increase of dentition, vacuolar degeneration of prickle cell layer and increase mitotic activity of basal cell layer together with dermal proliferation. Conclusion Under the condition of the present study, PDT using nanocurcumin photosensitizer was effective in the treatment of OSCC regarding clinical, histological and gene expression of BCL2 and Caspase-3.
Collapse
Affiliation(s)
- Latifa Mohamed Abdelgawad
- Department of Medical Applications of Lasers, National Institute of Laser Enhanced Sciences, Cairo University, Cairo, Egypt.
- Corresponding author: Latifa Mohamed Abdelgawad; Tel: +20 1005215402; E-mail:
| | - Ahmed Abdullatif Abdelaziz
- Department of Medical Applications of Lasers, National Institute of Laser Enhanced Sciences, Cairo University, Cairo, Egypt.
| | | | - Ali Mohamed Saafan
- Department of Medical Applications of Lasers, National Institute of Laser Enhanced Sciences, Cairo University, Cairo, Egypt.
| |
Collapse
|
8
|
Zhang D, Wang Y, Liu L, Li Z, Yang S, Zhao W, Wang X, Liao H, Zhou S. Establishment and evaluation of ectopic and orthotopic prostate cancer models using cell sheet technology. Lab Invest 2022; 20:381. [PMID: 36038939 PMCID: PMC9422158 DOI: 10.1186/s12967-022-03575-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/04/2022] [Indexed: 08/30/2023]
Abstract
Background The traditional prostate cancer (PCa) model is established by injecting cell suspension and is associated with a low tumor formation rate. Cell sheet technology is one of the advancements in tissue engineering for 3D cell-based therapy. In this study, we established ectopic and orthotopic PCa models by cell sheet technology, and then compared the efficiency of tumor formation with cell suspension injection. Methods DU145 cells were seeded on 35 mm temperature-sensitive dishes to form PCa cell sheets, while the cell suspension with the same cell density was prepared. After transplanting into the nude mice, the tumor volumes were measured every 3 days and the tumor growth curves were conducted. At the time points of 2 weeks and 4 weeks after the transplantation, magnetic resonance imaging (MRI) was used to evaluate the transplanting site and distant metastasis. Finally, the mice were sacrificed, and the related tissues were harvested for the further histological evaluation. Results The orthotopic tumor formation rate of the cell sheet injection group was obviously better than that in cell suspension injection group (100% vs 67%). Compared with cell suspension injection, the tumors of DU145 cell sheet fragments injection had the higher density of micro-vessels, more collagen deposition, and lower apoptosis rate. There was no evidence of metastasis in forelimb, lung and liver was found by MRI and histological tests. Conclusion We successfully cultured the DU145 cell sheet and can be used to establish ectopic and orthotopic PCa tumor-bearing models, which provide an application potential for preclinical drug development, drug-resistance mechanisms and patient individualized therapy.
Collapse
Affiliation(s)
- Dongliang Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Wang
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Lei Liu
- Department of Urology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Zeng Li
- Department of Urology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Shengke Yang
- Department of Urology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Weixin Zhao
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston Salem, NC, 27157, USA
| | - Xiang Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Liao
- Department of Urology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, China.
| | - Shukui Zhou
- Department of Urology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, China.
| |
Collapse
|
9
|
Haak VM, Huang S, Panigrahy D. Debris-stimulated tumor growth: a Pandora's box? Cancer Metastasis Rev 2021; 40:791-801. [PMID: 34665387 PMCID: PMC8524220 DOI: 10.1007/s10555-021-09998-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/01/2021] [Indexed: 12/24/2022]
Abstract
Current cancer therapies aim at eradicating cancer cells from the body. However, killing cells generates cell “debris” which can promote tumor progression. Thus, therapy can be a double-edged sword. Specifically, injury and debris generated by cancer therapies, including chemotherapy, radiation, and surgery, may offset their benefit by promoting the secretion of pro-tumorigenic factors (e.g., eicosanoid-driven cytokines) that stimulate regrowth and metastasis of surviving cells. The debris produced by cytotoxic cancer therapy can also contribute to a tumor microenvironment that promotes tumor progression and recurrence. Although not well understood, several molecular mechanisms have been implicated in debris-stimulated tumor growth that we review here, such as the involvement of extracellular vesicles, exosomal miR-194-5p, Bax, Bak, Smac, HMGB1, cytokines, and caspase-3. We discuss the cases of pancreatic and other cancer types where debris promotes postoperative tumor recurrence and metastasis, thus offering a new opportunity to prevent cancer progression intrinsically linked to treatment by stimulating resolution of tumor-promoting debris.
Collapse
Affiliation(s)
- Victoria M Haak
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Sui Huang
- Institute for Systems Biology, Seattle, WA, USA
| | - Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Patki M, Saraswat A, Bhutkar S, Dukhande V, Patel K. In vitro assessment of a synergistic combination of gemcitabine and zebularine in pancreatic cancer cells. Exp Cell Res 2021; 405:112660. [PMID: 34048785 DOI: 10.1016/j.yexcr.2021.112660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/13/2021] [Accepted: 05/12/2021] [Indexed: 01/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers with an extremely poor prognosis. Gemcitabine (Gem) is still the mainstay drug for the treatment of PDAC. However, rapid inactivation by cytidine deaminase (CDA) present in pancreatic cancer cells severely limits anticancer efficacy of Gem. In this study, we investigated the effect of a CDA inhibitor - Zebularine (Zeb) on anticancer activity of Gem in pancreatic cancer cell lines MiaPaCa-2, BxPC-3, and Panc-1. Zeb treatment synergistically increased Gem-induced cytotoxicity in all three pancreatic cancer cell lines. The strongest synergistic activity was found at 1:10 M ratio of Gem/Zeb (combination index 0.04-0.4). Additionally, Gem + Zeb treated cells showed marked decreased in the expressions of anti-apoptotic protein including Bcl-2 and survivin while significantly increased the cleaved caspase-3, and loss of mitochondrial membrane potential was observed. Multicellular 3D spheroids of MiaPaCa-2 cells treated with combination showed significant reduction (25-60%) in spheroid size, weight compared to single drug and control group. Live/dead cell imaging showed that Gem + Zeb treated spheroids exhibited a highly distorted surface with significantly higher number of dead cells (red). The results of the present study confirm that this synergistic combination is worthy of future investigations as a potential approach for the treatment of PDAC.
Collapse
Affiliation(s)
- Manali Patki
- College of Pharmacy and Health Sciences, St. John's University, NY, USA
| | | | - Shraddha Bhutkar
- College of Pharmacy and Health Sciences, St. John's University, NY, USA
| | - Vikas Dukhande
- College of Pharmacy and Health Sciences, St. John's University, NY, USA
| | - Ketan Patel
- College of Pharmacy and Health Sciences, St. John's University, NY, USA.
| |
Collapse
|
11
|
Asadi M, Taghizadeh S, Kaviani E, Vakili O, Taheri-Anganeh M, Tahamtan M, Savardashtaki A. Caspase-3: Structure, function, and biotechnological aspects. Biotechnol Appl Biochem 2021; 69:1633-1645. [PMID: 34342377 DOI: 10.1002/bab.2233] [Citation(s) in RCA: 142] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 08/01/2021] [Indexed: 12/16/2022]
Abstract
Caspase-3, a cysteine-aspartic acid protease, has recently attracted much attention because of its incredible roles in tissue differentiation, regeneration, and neural development. This enzyme is a key zymogen in cell apoptosis and is not activated until it is cleaved by initiator caspases during apoptotic flux. Since caspase-3 has represented valuable capabilities in the field of medical research, biotechnological aspects of this enzyme, including the production of recombinant type, protein engineering, and designing delivery systems, have been considered as emerging therapeutic strategies in treating the apoptosis-related disorders. To date, several advances have been made in the therapeutic use of caspase-3 in the management of some diseases such as cancers, heart failure, and neurodegenerative disorders. In the current review, we intend to discuss the caspase-3's structure, functions, therapeutic applications, as well as its molecular cloning, protein engineering, and relevant delivery systems.
Collapse
Affiliation(s)
- Marzieh Asadi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Taghizadeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elina Kaviani
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mortaza Taheri-Anganeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahshid Tahamtan
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
He B, Zhu Z, Chen F, Zhang R, Chen W, Zhang T, Wang T, Lei J. Synthesis and antitumor potential of new arylidene ursolic acid derivatives via caspase-8 activation. Arch Pharm (Weinheim) 2021; 354:e2000448. [PMID: 33646592 DOI: 10.1002/ardp.202000448] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/22/2021] [Accepted: 01/29/2021] [Indexed: 12/11/2022]
Abstract
Continuing our studies on NO-donating ursolic acid-benzylidene derivatives as potential antitumor agents, we designed and synthesized a series of new arylidene derivatives containing NO-donating ursolic acid and aromatic heterocyclic units. Compounds 5c and 6c showed a significant broad-spectrum antitumor activity. Compound 5c exhibited nearly three- to nine-fold higher cytotoxicity as compared with the parent drug in A549, MCF-7, HepG-2, HT-29, and HeLa cells, and it was also found to be the most potent apoptosis inducer of MCF-7 cells. More importantly, compound 5c arrested the MCF-7 cell cycle in the G1 phase, which was associated with caspase activation and a decrease of the Bcl-2/Bax ratio. Meanwhile, compound 5c caused changes in morphological features, dissipation of the mitochondrial membrane potential, and accumulation of reactive oxygen species. A docking study revealed that the nitroxyethyl moiety of compound 5c may form hydrogen bonds with caspase-8 amino acid residues (SER256 and HIS255). Together, these data suggest that NO-donating ursolic acid-arylidene derivatives are potent apoptosis inducers in tumor cells.
Collapse
Affiliation(s)
- Baoen He
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zuchang Zhu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fenglian Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rong Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiqiang Chen
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Te Zhang
- Department of Research and Development, Shanghai Hequan Pharmaceutical Co. Ltd., Shanghai, China
| | - Tao Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiamei Lei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
13
|
Khan I, Ganapathi T, Rehman MMU, Shareef MA, Kumar CG, Kamal A. New indenopyrazole linked oxadiazole conjugates as anti-pancreatic cancer agents: Design, synthesis, in silico studies including 3D-QSAR analysis. Bioorg Med Chem Lett 2021; 44:128094. [PMID: 33964437 DOI: 10.1016/j.bmcl.2021.128094] [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: 03/01/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 11/25/2022]
Abstract
To continue the quest of newer anticancer agents, herein a novel class of 1,4-Dihydroindenopyrazole linked oxadiazole conjugates 9(a-r) was designed, synthesized and experimented for their anti-proliferative activities against four different cancer cell lines (human) such as MDA MB-231 (breast), PANC-1 (pancreatic), MCF-7 (breast), and Caco-2 (Colorectal) by using MTT assay. Among the series compound 9h and 9 m demonstrated significant potency against the PANC-1 (human pancreatic cancer cells) with IC50 value 7.4 μM and 4.3 μM respectively. While compound 9 m was found to be equipotent to standard Gomitabine (IC50 = 4.2 μM). The detailed biological assays revealed S phase cell cycle arrest and their ability to propagate apoptosis by activating caspase 3 and 9 enzymes which was confirmed by Annexin-FITC assay and caspase assay. Moreover, docking study suggested their binding modes and interactions with caspase-3. In addition, in silico studies revealed that they exhibit good pharmacokinetics and drug likeliness properties. Furthermore, 3D-QSAR was carried out to achieve a pharmacophoric model with CoMFA (q2 = 0.631, r2 = 0.977) and CoMSIA (q2 = 0.686, r2 = 0.954) on PANC-1 cancer cells which were established, generated and validated to be reliable models for further design and optimization of newer molecules with enhanced anticancer activity.
Collapse
Affiliation(s)
- Irfan Khan
- Organic Synthesis and Process Chemistry Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India.
| | - Thipparapu Ganapathi
- Stem Cell Research Division, National Institute of Nutrition (NIN), Indian Council of Medical Research (ICMR), Hyderabad 500007, Telangana, India.
| | - Md Muzaffar-Ur- Rehman
- Catalysis and Fine Chemicals Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, India.
| | - Mohd Adil Shareef
- Academy of Scientific and Innovative Research, Ghaziabad 201002, India; Centre for Fluoro-Agrochemicals, CSIR-Indian Institute of Chemical Technology, Tarnaka 500007, Hyderabad, India.
| | - C Ganesh Kumar
- Organic Synthesis and Process Chemistry Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Ahmed Kamal
- Organic Synthesis and Process Chemistry Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India; School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
14
|
Tiliroside as a CAXII inhibitor suppresses liver cancer development and modulates E2Fs/Caspase-3 axis. Sci Rep 2021; 11:8626. [PMID: 33883691 PMCID: PMC8060393 DOI: 10.1038/s41598-021-88133-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 03/10/2021] [Indexed: 12/29/2022] Open
Abstract
Liver cancer is the fatal cause of cancer deaths worldwide due to its aggressiveness and lack of effective therapies. Tiliroside (C30H26O13) is an active compound extracted from herb plant Tribulus terrestris L., which has been used as alternative therapy in clinic practice. However, its therapeutic use against liver cancer has not been previously reported. Here, we showed that Tiliroside exerted significantly higher anti-proliferation effect on liver cancer cell lines Hep3B and SNU-449 than on liver normal cell THLE-3 cells or NC group, respectively, by using MTS assay. Results from colony formation, immigration and invasion assays support the anticancer efficacy of Tiliroside and its low-toxic property while treating liver normal cell THLE-3. 3D spheroid formation and CD133 expression level also displays its anti-stemness effect. It has been showed that Tiliroside may function as Carbonic anhydrases XII (CAXII) inhibitor and affects apoptotic E2F1/E2F3/Caspase-3 axis by using CAXII esterase activity assay, Human carbonic anhydrase 12 (CA-12) ELISA Kit, quantitative reverse transcription PCR (RT-qPCR) as well as CaspACE Assay System, respectively. In summary, we demonstrate for the first time that Tiliroside suppresses liver cancer development possibly by acting as a novel CAXII inhibitor, which warrant further investigation on its therapeutic implications.
Collapse
|
15
|
Iyappan P, Bala MD, Sureshkumar M, Veeraraghavan VP, Palanisamy A. D-carvone induced ROS mediated apoptotic cell death in human leukemic cell lines (Molt-4). Bioinformation 2021; 17:171-180. [PMID: 34393434 PMCID: PMC8340696 DOI: 10.6026/97320630017171] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 12/31/2020] [Accepted: 01/26/2021] [Indexed: 12/21/2022] Open
Abstract
The immature lymphoid cells with chromosomal structural and numerical abnormalities cause the acute lymphoblastic leukemia (ALL). This hematologic disorder constitutes about 25% of cancer prognosis among children and adolescents. D-Carvone, a monocyclic monoterpene obtained from the essential oils extracted from plants is reported to possess the various biological activities. The present study was aimed to investigate the anticancer potential of D-Carvone against the human leukemic Molt-4 cells. The cytotoxicity of DCarvone was analyzed by MTT assay. The level of lipid peroxidation and antioxidants were determined. The intracellular ROS, MMP and apoptosis were demonstrated by fluorescent staining techniques. The MTT assay revealed that the D-Carvone treatment suppressed the viability of Molt-4 cells and the IC50 was determined at 20 µM/ml. The D-Carvone treatment was increased the oxidative stress and reduced the level of antioxidants in the Molt-4 cell lines. The increased intracellular ROS, apoptotic cell death, and diminished MMP was noted in the D-Carvone treatment. In the Molt-4 cells, D-carvone induced the apoptosis in a time and dose dependent manner by the activation of caspases-8, -9 and -3. Thus, data provide insights for the clinical application of D-Carvone in the treatment of blood cancer Molt-4 cells. Our study suggests the therapeutic potential D-Carvone for the treatment of leukemia in future.
Collapse
Affiliation(s)
- Petchi Iyappan
- Senior Lecturer, Faculty of Medicine, Bioscience and Nursing, School of Bioscience, Mahsa University, Saujana Putra Campus, Jalan SP2, Bandar Saujana Putra, 42610, Jenjarom, Selangor, Malaysia
| | - M Devi Bala
- Research Scholar, Muthayammal College of Arts & Science (A Unit of VANETRA Group), Rasipuram, 637408, Namakkal, Tamilnadu, India
| | - M Sureshkumar
- Department of Zoology & Biotechnology, Muthayammal College of Arts & Science (A Unit of VANETRA Group), Rasipuram, 637408, Namakkal, Tamilnadu, India
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai - 600 077
| | - Arulselvan Palanisamy
- Adjunct Associate Professor,Muthayammal Centre for Advanced Research (MCAR), Muthayammal College of Arts & Science (A Unit of VANETRA Group),Rasipuram, 637408, Namakkal, Tamilnadu, India
| |
Collapse
|
16
|
A decade in unravelling the etiology of gastric carcinogenesis in Kashmir, India – A high risk region. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
17
|
Ghosh A, Chatterjee K, Chowdhury AR, Barui A. Clinico-pathological significance of Drp1 dysregulation and its correlation to apoptosis in oral cancer patients. Mitochondrion 2020; 52:115-124. [PMID: 32169612 DOI: 10.1016/j.mito.2020.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 01/06/2020] [Accepted: 03/09/2020] [Indexed: 12/14/2022]
Abstract
Dysregulation in mitochondrial dynamics has been associated with several diseases including cancer. Present study assessed the alteration in mitochondrial fission protein (Drp1) in oral epithelial cells collected from clinically confirmed pre-cancer and cancer patients and further correlates it with the cellular apoptosis signaling. Results indicate the ROS accumulation in OSCC patients is accompanied by several changes including increase in mitochondrial mass, expression of mitochondrial fission protein (Drp1) and alteration in apoptotic signaling. The positive co-relation has been observed between the expressions of anti-apoptotic Bcl-2proteinswith mitochondrial fission protein Drp1. Higher mitochondrial fission in oral cancer cells was also correlated with the increased expression of cell cycle marker CyclinD1 indicating highly proliferative stage of oral cancer cells. The clinical correlation can be extended to develop biomarker for diagram and program in oral cancer management.
Collapse
Affiliation(s)
- Aritri Ghosh
- Centre for Healthcare Science and Technology, Indian Institute of Engineering, Science and Technology, P.O. Botanic Garden, Shibpur, Howrah 711103, WB, India
| | - Kabita Chatterjee
- Department of Oral and Maxillofacial Pathology, Buddha Institute of Dental Sciences, West of TV Tower, Gandhinagar, Kankarbagh, Patna 800020, Bihar, India
| | - Amit Roy Chowdhury
- Department of Aerospace and Applied Mechanics, Indian Institute of Engineering Science and Technology, Shibpur, P.O. Botanic Garden, Shibpur, Howrah 711103, WB, India
| | - Ananya Barui
- Centre for Healthcare Science and Technology, Indian Institute of Engineering, Science and Technology, P.O. Botanic Garden, Shibpur, Howrah 711103, WB, India.
| |
Collapse
|
18
|
Caspase-8: The double-edged sword. Biochim Biophys Acta Rev Cancer 2020; 1873:188357. [PMID: 32147543 DOI: 10.1016/j.bbcan.2020.188357] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/13/2020] [Accepted: 03/03/2020] [Indexed: 12/17/2022]
Abstract
Caspase-8 is a cysteine - aspartate specific protease that classically triggers the extrinsic apoptotic pathway, in response to the activation of cell surface Death Receptors (DRs) like FAS, TRAIL-R and TNF-R. Besides it's roles in triggering death receptor-mediated apoptosis, Caspase-8 has also been implicated in the onsets of anoikis, autophagy and pyroptosis. Furthermore, Caspase-8 also plays a crucial pro-survival function by inhibiting an alternative form of programmed cell death called necroptosis. Low expression levels of pro-Caspase-8 is therefore associated with the malignant transformation of cancers. However, the long-held notion that pro-Caspase-8 expression/activity is generally lost in most cancers, thereby contributing to apoptotic escape and enhanced resistance to anti-cancer therapeutics, has been found to be true for only a minority of cancers types. In the majority of cases, pro-Caspase-8 expression is maintained and sometimes elevated, while it's apoptotic activity is regulated through different mechanisms. This supports the notion that the non-apoptotic functions of Caspase-8 offer growth advantage in these cancer types and have, therefore, gained renewed interest in the recent years. In light of these reasons, a number of therapeutic approaches have been employed, with the intent of targeting pro-Caspase-8 in cancer cells. In this review, we would attempt to discuss - the classic roles of Caspase-8 in initiating apoptosis; it's non-apoptotic functions; it's the clinical significance in different cancer types; and the therapeutic applications exploiting the ability of pro-Caspase-8 to regulate various cellular functions.
Collapse
|
19
|
Yang M, Qin Q, Zhu J, Guo Y, Yin T, Wu H, Wang C. Long noncoding RNA ITGB2-AS1 promotes growth and metastasis through miR-4319/RAF1 axis in pancreatic ductal adenocarcinoma. J Cell Physiol 2020. [PMID: 31957875 DOI: 10.1002/jcp.29471] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 01/08/2020] [Indexed: 12/11/2022]
Abstract
Long noncoding RNA (lncRNA) has been considered as potentially critical regulators in pancreatic ductal adenocarcinoma (PDAC). In this study, we prospectively investigate the effect and mechanism of lncRNA integrin subunit beta 2-anti-sense RNA 1 (ITGB2-AS1) on regulation of PDAC progression. The expression of ITGB2-AS1 and its target were analyzed by quantitative real-time polymerase chain reaction and in situ hybridization. 3-(4,5-Dimethylthiazol-z-yl)-2,5-diphenyltetrazolium bromide, flow cytometry, wound healing, and transwell assays were used to investigate the influence of ITGB2-AS1 on cell proliferation, cell cycle, migration, and invasion, respectively. The interaction between ITGB2-AS1 and its target was determined via luciferase activity assay and RNA immunoprecipitation. The subcutaneous xenotransplanted tumor model was established and employed to detect the tumorigenic function of ITGB2-AS1, which was evaluated by western blot analysis, immunohistochemistry, and hematoxylin and eosin staining. The results showed that ITGB2-AS1 was elevated in both PDAC tumor tissues and cell lines, predicting a poor prognosis in PDAC patients. Knocking down of ITGB2-AS1 suppressed PDAC cell proliferation, invasion, and migration but induced cell apoptosis in vitro. Moreover, ITGB2-AS1 could target and inhibit the expression of miR-4319 and miR-4319-targeted and -suppressed serine/threonine kinase RAF1. ITGB2-AS1 promoted PDAC progression via inhibition of miR-4319. Interference of ITGB2-AS1 could suppress in vivo tumorigenic ability of PDAC via downregulation of RAF1. In conclusion, ITGB2-AS1 promoted PDAC progression via sponging miR-4319 to upregulate RAF1, suggesting the potential therapeutic target ability of ITGB2-AS1 in PDAC.
Collapse
Affiliation(s)
- Ming Yang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qi Qin
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Junling Zhu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yao Guo
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Yin
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Heshui Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chunyou Wang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
20
|
Boudreau MW, Peh J, Hergenrother PJ. Procaspase-3 Overexpression in Cancer: A Paradoxical Observation with Therapeutic Potential. ACS Chem Biol 2019; 14:2335-2348. [PMID: 31260254 PMCID: PMC6858495 DOI: 10.1021/acschembio.9b00338] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many anticancer strategies rely on the promotion of apoptosis in cancer cells as a means to shrink tumors. Crucial for apoptotic function are executioner caspases, most notably caspase-3, that proteolyze a variety of proteins, inducing cell death. Paradoxically, overexpression of procaspase-3 (PC-3), the low-activity zymogen precursor to caspase-3, has been reported in a variety of cancer types. Until recently, this counterintuitive overexpression of a pro-apoptotic protein in cancer has been puzzling. Recent studies suggest subapoptotic caspase-3 activity may promote oncogenic transformation, a possible explanation for the enigmatic overexpression of PC-3. Herein, the overexpression of PC-3 in cancer and its mechanistic basis is reviewed; collectively, the data suggest the potential for exploitation of PC-3 overexpression with PC-3 activators as a targeted anticancer strategy.
Collapse
Affiliation(s)
- Matthew W. Boudreau
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| | - Jessie Peh
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| | - Paul J. Hergenrother
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| |
Collapse
|
21
|
Jia HY, Zhang K, Lu WJ, Xu GW, Zhang JF, Tang ZL. LncRNA MEG3 influences the proliferation and apoptosis of psoriasis epidermal cells by targeting miR-21/caspase-8. BMC Mol Cell Biol 2019; 20:46. [PMID: 31660855 PMCID: PMC6816225 DOI: 10.1186/s12860-019-0229-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/23/2019] [Indexed: 12/14/2022] Open
Abstract
Background It was reported that microRNA-21(miR-21) was differentially expressed in the keratinocytes of psoriasis patients, and it may influence the apoptosis and proliferation of cells. The role of lncRNA maternally expressed gene3 (MEG3), a competing endogenous RNAs of miR-21, in the progression of psoriasis remains unclear. We aimed to unfold the influence of MEG3 and miR-21 on the proliferation and apoptosis of psoriasis epidermal cells. Methods 50μg/L TNF-α was used to treat HaCaTs and NHEKs cells for 24 h, and then different experiments were conducted. qRT-PCR were applied for measuring the mRNA level of MEG3, miR-2, and caspase-8, and the protein expression of caspase-8 was measured with western blotting. Flow cytometry was used for assessing apoptosis. Cell proliferation was detected using MTT and colony formation assays. Dual luciferase reporter assay was applied for confirming the binding site between MEG3 and miR-21, miR-21 and Caspase-8. Results A cell model for in vitro studying the role of MEG3 in psoriasis pathophysiology was established using HaCaT and HHEKs. MEG3 was significantly down-regulated in HaCaT, HHEKs, and psoriatic skin samples. MEG3 inhibits proliferation and promotes apoptosis of Activated-HaCaT (Act-HaCaT) and Activated-HHEKs (Act- HHEK) by regulating miR-21, and the binding site between MEG3 and miR-21 was identified. We also found that miR-21 could inhibit the level of caspase-8 and identified the binding site between caspase-8 and miR-21. Some down-stream proteins of caspase-8, Cleaved caspase-8, cytc, and apaf-1 were regulated by miR-21 and MEG3. Conclusion MEG3/miR-21 axis may regulate the expression of caspase-8, and further influence the proliferation and apoptosis of psoriasis keratinocyte, Act-HaCaT and Act- HHEK. Therefore, our findings may provide a new thought for the study of pathogenesis and treatment of psoriasis.
Collapse
Affiliation(s)
- Hai-Yan Jia
- Department of Dermatology, Qilu Hospital of Shandong University, No.107, West Wenhua Road, Jinan, 250012, Shandong Province, People's Republic of China
| | - Kai Zhang
- Department of Neurosurgery, Shengli Oilfield Central Hospital, Dongying, 257000, People's Republic of China
| | - Wen-Jing Lu
- Department of Dermatology, Qilu Hospital of Shandong University, No.107, West Wenhua Road, Jinan, 250012, Shandong Province, People's Republic of China
| | - Gui-Wen Xu
- Department of Dermatology, Qilu Hospital of Shandong University, No.107, West Wenhua Road, Jinan, 250012, Shandong Province, People's Republic of China
| | - Jian-Fen Zhang
- Department of Dermatology, Qilu Hospital of Shandong University, No.107, West Wenhua Road, Jinan, 250012, Shandong Province, People's Republic of China
| | - Zhan-Li Tang
- Department of Dermatology, Qilu Hospital of Shandong University, No.107, West Wenhua Road, Jinan, 250012, Shandong Province, People's Republic of China.
| |
Collapse
|
22
|
Han R, Chen X, Li Y, Zhang S, Li R, Lu L. MicroRNA-34a suppresses aggressiveness of hepatocellular carcinoma by modulating E2F1, E2F3, and Caspase-3. Cancer Manag Res 2019; 11:2963-2976. [PMID: 31114344 PMCID: PMC6489561 DOI: 10.2147/cmar.s202664] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/04/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Accumulating evidence suggests an antineoplastic role of MicroRNA-34a (miR-34a) in human cancer. However, its precise biological functions stay largely elusive. Purpose: Our study was aimed to investigate the impact of miR-34a on hepatocellular carcinoma (HCC) and its underlying apoptosis related mechanisms in vitro, as well as the association of miR-34a, E2F1 and E2F3 expression with patient survival of HCC using publicly accessed datasets. Methods: The HBV-expressing Hep3B and SNU-449 cell lines with or without enforced expression of miR-34a were in vitro cultured for cell proliferation, colony formation, wound healing, cell invasion, and 3D spheroid formation. Quantitative reverse transcription PCR (RT-qPCR) was performed for E2F1, E2F3 expression. Caspase-3 (CASP3) activity was determined using a CaspACETM Assay System. Kaplan-Meier survival curves were used to analyze the associations of miR-34a, E2F1 and E2F3 expression and overall survival in HCC. Meta-analysis was performed to examine the differential expression of E2F1 and E2F3 between primary HCC vs normal tissues. Results: The results in vitro showed that enforced miR-34a expression significantly inhibited cell proliferation, migration, and invasion of both Hep3B and SNU-449. RT-qPCR results demonstrated that miR-34a could significantly suppress E2F1 and E2F3 expression, particularly in SNU-449. CASP3 activity in both Hep3B and SNU-449 increased in miR-34a treatment group. Overexpressed E2F1 and E2F3 were observed in primary HCC vs normal tissues. Survival analyses showed that HCC patients with either high miR-34a, or low E2F1, or low E2F3 expression had better survival than their opposite counterparts, respectively. Conclusion: Our study suggested thatmiR-34a can modulate the expression of E2F1, E2F3, and CASP3 activity, thereby repressing tumor aggressiveness and expediting apoptosis in liver cancer cells.
Collapse
Affiliation(s)
- Rui Han
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700 People's Republic of China.,Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xinyi Chen
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700 People's Republic of China
| | - Ya Li
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700 People's Republic of China.,Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Shunjia Zhang
- Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ruibai Li
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700 People's Republic of China
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, School of Medicine, Yale University, New Haven, CT, 06520-8034, USA.,Center for Biomedical Data Science, Yale Cancer Center, Yale University, New Haven, CT, USA
| |
Collapse
|
23
|
Mitochondria-targeting BODIPY-loaded micelles as novel class of photosensitizer for photodynamic therapy. Eur J Med Chem 2018; 157:599-609. [PMID: 30125721 DOI: 10.1016/j.ejmech.2018.08.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/23/2018] [Accepted: 08/09/2018] [Indexed: 11/22/2022]
Abstract
In this paper, a series of novel BODIPY-based photosensitizers have been designed and synthesized for photodynamic therapy. BODIPY3 was screened out as the most potential photosensitizer due to its excellent optical properties, high singlet oxygen efficiency and good photostability. However, as an organic photosensitizer, BODIPY3 still suffered from the drawbacks of insolubility and instability in aqueous system. In view of these problems, DSPE-PEG2000 was used to trap BODIPY3 into the hydrophobic core of micelles to obtain well-dispersing nano complexes BODIPY3-PEG3 in aqueous system. More importantly, BODIPY3-PEG3 not only has better solubility and stability in aqueous media but can generate significant singlet oxygen (1O2, one of the reactive oxygen species, the real cytotoxic agent in photodynamic therapy) in living cells and exhibit high light cytotoxicity to three cancer cell lines. The mechanism studies indicated the mitochondrial localization of BODIPY3-PEG3 was able to generate ROS in mitochondria, which further result in mitochondrial dysfunction and photoinduced apoptosis via caspase-8 and caspase-3 pathway.
Collapse
|
24
|
Huang JS, Yang CM, Wang JS, Liou HH, Hsieh IC, Li GC, Huang SJ, Shu CW, Fu TY, Lin YC, Ger LP, Liu PF. Caspase-3 expression in tumorigenesis and prognosis of buccal mucosa squamous cell carcinoma. Oncotarget 2017; 8:84237-84247. [PMID: 29137419 PMCID: PMC5663591 DOI: 10.18632/oncotarget.20494] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 07/26/2017] [Indexed: 12/18/2022] Open
Abstract
Buccal mucosa squamous cell carcinoma (BMSCC) is the most common oral cancer in Southeast Asia. Caspase-3, a key molecule in regulating apoptosis, promotes the malignancy of various cancers. However, its role in BMSCC is unknown. Herein, we evaluated the association of caspase-3 expression with tumorigenesis and prognosis in BMSCC patients. Immunohistochemical staining indicated that the expression levels of cleaved caspase-3 (p<0.001) and caspase-3 (p<0.001) in 185 BMSCC tissues were significantly higher compared to those in the tumor adjacent normal tissues. Moreover, the high expression of caspase-3 was associated with poor pathological outcomes [advanced pathological stage (p=0.029) and larger tumor size (p=0.002)] and poor disease-free survival in patients receiving postoperative radiotherapy (p=0.030). Moreover, the low co-expression of cleaved caspase-3 and caspase-3 was associated with better disease-specific survival in patients with early pathological stage (I + II, p=0.018) or without lymph node invasion (p=0.043) compared to the positive/high expression of either or both cleaved caspase-3 and caspase-3. Taken together, cleaved caspase-3 and caspase-3 could be biomarkers for tumorigenesis in BMSCC patients. Cleaved caspase-3 and/or caspase-3 might be prognostic biomarkers for certain stages of BMSCC.
Collapse
Affiliation(s)
- Jer-Shyung Huang
- Department of Radiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Cheng-Mei Yang
- Department of Stomatology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Department of Dental Technology, Shu-Zen Junior College of Medicine and Management, Kaohsiung, Taiwan
| | - Jyh-Seng Wang
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Huei-Han Liou
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - I-Chien Hsieh
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Guan-Cheng Li
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Sin-Jhih Huang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chih-Wen Shu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Ting-Ying Fu
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yun-Chung Lin
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan
| | - Luo-Ping Ger
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Pei-Feng Liu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Department of Optometry, Shu-Zen Junior College of Medicine and Management, Kaohsiung, Taiwan
| |
Collapse
|
25
|
Xu B, Xia H, Cao J, Wang Z, Yang Y, Lin Y. MicroRNA-21 Inhibits the Apoptosis of Osteosarcoma Cell Line SAOS-2 via Targeting Caspase 8. Oncol Res 2017; 25:1161-1168. [PMID: 28109080 PMCID: PMC7841250 DOI: 10.3727/096504017x14841698396829] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Currently, multiple microRNAs (miRNAs) have been found to play vital roles in the pathogenesis of osteosarcoma. This study aimed to investigate the role of miR-21 in osteosarcoma. The level of miR-21 in 20 pairs of osteosarcoma and corresponding adjacent tissues was monitored by qPCR. Human osteosarcoma cell line SAOS-2 was transfected with either miR-21 mimic or miR-21 inhibitor, and then cell viability, survival, and apoptosis were measured by MTT, colony formation assay, and flow cytometry. A target of miR-21 was predicted by the microRNA.org database and verified in vitro by using luciferase reporter, qPCR, and Western blot analyses. Finally, cells were cotransfected with siRNA against caspase 8 and miR-21 inhibitor, and the apoptotic cell rate was determined again. Results showed that the mRNA level of miR-21 was highly expressed in osteosarcoma tissues compared with adjacent tissues. Overexpression of miR-21 improved cell viability and survival but suppressed apoptosis. Caspase 8 was a direct target of miR-21, and it was negatively regulated by miR-21. Moreover, miR-21 suppression attenuated caspase 8 silencing and induced the decrease in apoptosis. In conclusion, overexpression of miR-21 suppressed SAOS-2 cell apoptosis via directly targeting caspase 8.
Collapse
Affiliation(s)
- Bin Xu
- Department of Orthopedics, The Third Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Hehuan Xia
- Department of Orthopedics, The Third Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Junming Cao
- Department of Orthopedics, The Third Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Zhihong Wang
- Department of Orthopedics, The Third Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Yipeng Yang
- Department of Orthopedics, The Third Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Yongsheng Lin
- Department of Orthopedics, The Third Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| |
Collapse
|
26
|
Liu PF, Hu YC, Kang BH, Tseng YK, Wu PC, Liang CC, Hou YY, Fu TY, Liou HH, Hsieh IC, Ger LP, Shu CW. Expression levels of cleaved caspase-3 and caspase-3 in tumorigenesis and prognosis of oral tongue squamous cell carcinoma. PLoS One 2017; 12:e0180620. [PMID: 28700659 PMCID: PMC5503265 DOI: 10.1371/journal.pone.0180620] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/19/2017] [Indexed: 01/07/2023] Open
Abstract
Apoptosis plays a dual role in cancer development and malignancy. The role of apoptosis-related caspases in cancer remains controversial, particularly in oral tongue squamous cell carcinoma (OTSCC). In this study, we examined the protein levels of cleaved caspase-3, caspase-3, caspase-8, and caspase-9 on tissue microarrays consisting of samples from 246 OTSCC patients by immunohistochemistry. Wilcoxon signed-rank test indicated that the protein levels of cleaved caspase-3, caspase-3, caspase-8, and caspase-9 in tumor tissues were significantly higher compared to those in adjacent normal tissues (all p<0.001). The expression level of caspase-8 in tumors was elevated in patients with lymph node invasion. Moreover, positive expression of cleaved caspase-3 was associated with shorter disease-free survival (DFS) in OTSCC patients with moderate differentiation and lymph node invasion. Combination of either positive cleaved caspase-3 or higher caspase-3 expression or both was associated with poor DFS. Interestingly, stratification analysis showed that co-expression levels of positive cleaved caspase-3 or/and higher caspase-3 were associated with better disease-specific survival in patients with advanced stages of the disease, such as large tumor size and lymph node invasion, whereas it was associated with poor DFS in OTSCC patients with moderate cell differentiation and small tumor size. Taken together, cleaved caspase-3 and caspase-3/8/9 could be biomarkers for tumorigenesis in OTSCC patients. The co-expression level of cleaved caspase-3 and caspase-3 might be a prognostic biomarker for OTSCC patients, particular in those patients with certain tumor stages and cell differentiation status.
Collapse
Affiliation(s)
- Pei-Feng Liu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yu-Chang Hu
- Department of Radiation Oncology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Bor-Hwang Kang
- Department of Otorhinolaryngology-Head & Neck Surgery, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Otorhinolaryngology, School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Kai Tseng
- Department of Orthopedics, Show Chwan Memorial Hospital, Changhua, Taiwan
- Department of Orthopedics, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Pi-Chuang Wu
- Department of Nutrition, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chi-Chuang Liang
- Department of Nutrition, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yu-Yi Hou
- Department of Otorhinolaryngology-Head & Neck Surgery, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Ting-Ying Fu
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Huei-Han Liou
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - I-Chien Hsieh
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Luo-Ping Ger
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
- * E-mail: (CWS); (LPG)
| | - Chih-Wen Shu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- * E-mail: (CWS); (LPG)
| |
Collapse
|
27
|
Van den Bossche J, Deben C, Op de Beeck K, Deschoolmeester V, Hermans C, De Pauw I, Jacobs J, Van Schil P, Vermorken JB, Pauwels P, Peeters M, Lardon F, Wouters A. Towards Prognostic Profiling of Non-Small Cell Lung Cancer: New Perspectives on the Relevance of Polo-Like Kinase 1 Expression, the TP53 Mutation Status and Hypoxia. J Cancer 2017. [PMID: 28638459 PMCID: PMC5479250 DOI: 10.7150/jca.18455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Background: Currently, prognosis of non-small cell lung cancer (NSCLC) patients is based on clinicopathological factors, including TNM stage. However, there are considerable differences in patient outcome within a similar staging group, even when patients received identical treatments. In order to improve prognostic predictions and to guide treatment options, additional parameters influencing outcome are required. Polo-like kinase 1 (Plk1), a master regulator of mitotic cell division and the DNA damage response, is considered as a new potential biomarker in this research area. While several studies reported Plk1 overexpression in a broad range of human malignancies, inconsistent results were published regarding the clinical significance hereof. A prognostic panel, consisting of Plk1 and additional biomarkers that are related to the Plk1 pathway, might further improve prediction of patient prognosis. Methods: In this study, we evaluated for the first time the prognostic value of Plk1 mRNA and protein expression in combination with the TP53 mutation status (next generation sequencing), induction of apoptotic cell death (immunohistochemistry for cleaved caspase 3) and hypoxia (immunohistochemistry for carbonic anhydrase IX (CA IX)) in 98 NSCLC adenocarcinoma patients. Results: Both Plk1 mRNA and protein expression and CA IX protein levels were upregulated in the majority of tumor samples. Plk1 mRNA and protein expression levels were higher in TP53 mutant samples, suggesting that Plk1 overexpression is, at least partially, the result of loss of functional p53 (<0.05). Interestingly, the outcome of patients with both Plk1 mRNA and CA IX protein overexpression, who also harbored a TP53 mutation, was much worse than that of patients with aberrant expression of only one of the three markers (p=0.001). Conclusion: The combined evaluation of Plk1 mRNA expression, CA IX protein expression and TP53 mutations shows promise as a prognostic panel in NSCLC patients. Moreover, these results pave the way for new combination strategies with Plk1 inhibitors.
Collapse
Affiliation(s)
- Jolien Van den Bossche
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Christophe Deben
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.,Department of Pathology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Ken Op de Beeck
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.,Center of Medical Genetics, University of Antwerp, Antwerp University Hospital, Prins Boudewijnlaan 43, 2650 Edegem, Belgium
| | - Vanessa Deschoolmeester
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.,Department of Pathology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Christophe Hermans
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.,Department of Pathology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Ines De Pauw
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Julie Jacobs
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.,Department of Pathology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Paul Van Schil
- Department of Thoracic and Vascular Surgery, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Jan Baptist Vermorken
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.,Department of Oncology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Patrick Pauwels
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.,Department of Pathology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Marc Peeters
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.,Department of Oncology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Filip Lardon
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - An Wouters
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|