1
|
Fang XL, Cao XP, Xiao J, Hu Y, Chen M, Raza HK, Wang HY, He X, Gu JF, Zhang KJ. Overview of role of survivin in cancer: expression, regulation, functions, and its potential as a therapeutic target. J Drug Target 2024; 32:223-240. [PMID: 38252514 DOI: 10.1080/1061186x.2024.2309563] [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: 07/11/2023] [Accepted: 11/11/2023] [Indexed: 01/24/2024]
Abstract
Survivin holds significant importance as a member of the inhibitor of apoptosis protein (IAP) family due to its predominant expression in tumours rather than normal terminally differentiated adult tissues. The high expression level of survivin in tumours is closely linked to chemotherapy resistance, heightened tumour recurrence, and increased tumour aggressiveness and serves as a negative prognostic factor for cancer patients. Consequently, survivin has emerged as a promising therapeutic target for cancer treatment. In this review, we delve into the various biological characteristics of survivin in cancers and its pivotal role in maintaining immune system homeostasis. Additionally, we explore different therapeutic strategies aimed at targeting survivin.
Collapse
Affiliation(s)
- Xian-Long Fang
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
| | - Xue-Ping Cao
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
| | - Jun Xiao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yun Hu
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
| | - Mian Chen
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
| | - Hafiz Khuram Raza
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
| | - Huai-Yuan Wang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xu He
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jin-Fa Gu
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
| | - Kang-Jian Zhang
- Academician Expert Workstation of Fengxian District, Shanghai Yuansong Biotechnology Limited Company, Shanghai, China
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
2
|
Zhao B, Liang Z, Zhang L, Jiang L, Xu Y, Zhang Y, Zhang R, Wang C, Liu Z. Ponicidin Promotes Hepatocellular Carcinoma Mitochondrial Apoptosis by Stabilizing Keap1-PGAM5 Complex. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406080. [PMID: 39116422 PMCID: PMC11481384 DOI: 10.1002/advs.202406080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/09/2024] [Indexed: 08/10/2024]
Abstract
Ponicidin is a diterpenoid with demonstrated antitumor activity in clinical trials. However, the specific function and mechanism of action against hepatocellular carcinoma (HCC) remain unknown. In this study, it is found that ponicidin significantly inhibited the proliferation and migration of HCC cells. It is shown that ponicidin targets Keap1 and promotes the formation of the Keap1-PGAM5 complex, leading to the ubiquitination of PGAM5, using biotin-labeled ponicidin for target fishing and the HuProtTM Human Proteome Microarray V4.0. Ponicidin is found to activate the cysteine-dependent mitochondrial pathway via PGAM5, resulting in mitochondrial damage and ROS production, thereby promoting mitochondrial apoptosis in HepG2 cells. The first in vitro cocrystal structure of the PGAM5 IE 12-mer peptide and the Keap1 Kelch domain is obtained. Using molecular dynamics simulations to confirm the binding of ponicidin to the Keap1-PGAM5 complex. Based on the depth-based dynamic simulation, it is found that ponicidin can induce the tightening of the Keap1-PGAM5 interaction pocket, thereby stabilizing the formation of the protein complex. Finally, it is observed that ponicidin effectively inhibited tumor growth and promoted tumor cell apoptosis in a BALB/c nude mouse xenograft tumor model. The results provide insight into the anti-HCC properties of ponicidin based on a mechanism involving the Keap1-PGAM5 complex.
Collapse
Affiliation(s)
- Bixin Zhao
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Zuhui Liang
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Lisheng Zhang
- Research Center of Integrative MedicineSchool of Basic Medical ScienceGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Lin Jiang
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Yuanhang Xu
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Ying Zhang
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Rong Zhang
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Caiyan Wang
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Zhongqiu Liu
- State Key Laboratory of Traditional Chinese Medicine SyndromeInternational Institute for Translational Chinese MedicineGuangzhou University of Chinese MedicineGuangzhou510006China
| |
Collapse
|
3
|
Xu W, Hu J, Ma Z, Feng W, Gong W, Fu S, Chen X. Decreased BIRC5-206 promotes epithelial-mesenchymal transition in nasopharyngeal carcinoma through sponging miR-145-5p. Open Med (Wars) 2024; 19:20241007. [PMID: 39308922 PMCID: PMC11416051 DOI: 10.1515/med-2024-1007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/23/2024] [Accepted: 07/01/2024] [Indexed: 09/25/2024] Open
Abstract
Metastasis significantly contributes to the poor prognosis of advanced nasopharyngeal carcinoma (NPC). Our prior studies have demonstrated a decrease in BIRC5-206 expression in NPC, which promotes disease progression. However, the role of BIRC5-206 in the invasion and metastasis of NPC has not been fully elucidated. In this study, our objective was to explore the biological function and underlying mechanisms of BIRC5-206 in NPC. Additionally, we established an NPC mouse model of lung invasiveness using C666 cells to assess the impact of BIRC5-206 on NPC metastasis. Our results revealed that silencing BIRC5-206 inhibited apoptosis and enhanced the invasion of NPC cells, whereas its overexpression reversed these effects. Moreover, decreased BIRC5-206 expression significantly increased N-cadherin and Vimentin expression while reducing E-cadherin and occludin levels, both in vivo and in vitro. Additionally, silencing BIRC5-206 markedly augmented the formation of invasive foci in lung tissues. Rescue experiments further confirmed that decreased BIRC5-206 expression facilitates NPC metastasis via modulation of the miR-145-5p/CD40 signaling pathway. In summary, our study suggests that BIRC5-206 may serve as a potential prognostic biomarker and therapeutic target in the diagnosis and treatment of NPC.
Collapse
Affiliation(s)
- Weihua Xu
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Hainan Tropical Cancer Research Institute, Haikou, Hainan, 570312, China
| | - Junjie Hu
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Hainan Tropical Cancer Research Institute, Haikou, Hainan, 570312, China
| | - Zhichao Ma
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Hainan Tropical Cancer Research Institute, Haikou, Hainan, 570312, China
| | - Wanyi Feng
- Hainan Lvtou Medical Laboratory Center, Haikou, Hainan, 570206, China
- School of Life Sciences, Hainan University, Haikou, Hainan, 570228, China
| | - Wei Gong
- Hainan Lvtou Medical Laboratory Center, Haikou, Hainan, 570206, China
- School of Life Sciences, Hainan University, Haikou, Hainan, 570228, China
| | - Shengmiao Fu
- Central Laboratory, Hainan General Hospital, Hainan Hospital Affiliated to the Hainan Medical College, No. 19 Xiuhua Road, Xiuying District, Haikou, Hainan, 570311, China
- Hainan Lvtou Medical Laboratory Center, No. 16 Jinyu East Road, Longhua District, Haikou, Hainan, 570206, China
| | - Xinping Chen
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Hainan Tropical Cancer Research Institute, No. 6, Changbin West 4th Street, Xiuying District, Haikou, Hainan, 570312, China
| |
Collapse
|
4
|
Sun L, Jiao YW, Cui FQ, Liu J, Xu ZY, Sun DL. tRF-Leu reverse breast cancer cells chemoresistance by regulation of BIRC5. Discov Oncol 2024; 15:449. [PMID: 39278863 PMCID: PMC11402887 DOI: 10.1007/s12672-024-01317-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 09/05/2024] [Indexed: 09/18/2024] Open
Abstract
OBJECTIVE Accumulating studies reported the crucial roles of tRFs in tumorigenesis. However, their further mechanisms and clinical values remains unclear. This study aimed at the further investigation of tRF-Leu in breast cancer chemotherapy resistance. METHODS The high-throughput sequencing was performed and identified the downregulation of tRF-Leu in MCF7/ADR cells. The function of tRF-Leu in breast cancer cells and breast cancer chemotherapy resistance was investigated in vitro and in vivo, including colony formation assay, CCK-8 assay, transwell assay and apoptosis assay. The binding site of tRF-Leu on BIRC5 was verified by dual-luciferase assay. RESULTS tRF-Leu was downregulated in MCF7/ADR cells. Overexpression of tRF-Leu inhibited the migration of breast cancer cells. Furthermore, tRF-Leu could reverse the resistance of MCF7/ADR cells to Adriamycin both in vitro and in vivo. BIRC5 was a target of tRF-Leu, which might be involved in the chemotherapy resistance regulation. CONCLUSION We demonstrated that tRF-Leu could inhibit the chemotherapy resistance of breast cancer by targeting BIRC5. These findings might identify new biomarkers of breast cancer therapy and bring new strategies to reverse chemotherapy resistance.
Collapse
Affiliation(s)
- Li Sun
- Hepatopancreatobiliary Surgery Department, The Third Affiliated Hospital of Soochow University, Changzhou First People's Hospital, Changzhou, China
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yu-Wen Jiao
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Fu-Qi Cui
- Department Graduate School of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Jin Liu
- Department Graduate School of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Zhong-Ya Xu
- Children's Hospital of Nanjing Medical University, Nanjing, China.
| | - Dong-Lin Sun
- Hepatopancreatobiliary Surgery Department, The Third Affiliated Hospital of Soochow University, Changzhou First People's Hospital, Changzhou, China.
| |
Collapse
|
5
|
Awaji AA, Alhamdi HW, Alshehri KM, Alfaifi MY, Shati AA, Elbehairi SEI, Radwan NAF, Hafez HS, Elshaarawy RFM, Welson M. Bio-molecular Fe(III) and Zn(II) complexes stimulate the interplay between PI3K/AKT1/EGFR inhibition and induce autophagy and apoptosis in epidermal skin cell cancer. J Inorg Biochem 2024; 262:112720. [PMID: 39243420 DOI: 10.1016/j.jinorgbio.2024.112720] [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: 06/29/2024] [Revised: 08/20/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
This study investigated the effectiveness and safety of a hybrid thiosemicarbazone ligand (HL) and its metal complexes (MnII-L, FeIII-L, NiII-HL, and ZnII-HL) against epidermoid carcinoma (A-431). The results indicated that FeIII-L is the most effective, with a high selectivity index of 8.01 and an IC50 of 17.49 ± 2.12 μM for FeIII-L. The study also revealed that the synthesized complexes effectively inhibited gene expression of the Phosphoinositide 3-kinases (PI3K), alpha serine/threonine-protein kinase (AKT1), epidermal growth factor receptor (EGFR2) axis mechanism (P < 0.0001). Additionally, these complexes trigger a chain of events that include the inhibition of proliferating cell nuclear antigen (PCNA), transforming growth factor β1 (TGF β1), and topoisomerase II, and leading to a decrease in epidermoid cell proliferation. Furthermore, the inhibitory activity also resulted in the upregulation of caspases 3 and 9, indicating the acceleration of apoptotic markers, and the down regulation of miRNA221, suggesting a decrease in epidermoid proliferation. Molecular modeling of FeIII-L revealed that it had the best binding energy -8.02 kcal/mol and interacted with five hydrophobic π-interactions with Val270, Gln79, Leu210, and Trp80 against AKT1. Furthermore, the binding orientation of FeIII-L with Topoisomerase II was found to be the most stable, with a binding energy -8.25 kcal/mol. This stability was attributed to the presence of five hydrophobic π-interactions with His759, Guanin13, Cytosin8, and Ala465, and numerous ionic interactions, which were more favorable than those of doxorubicin and etoposide for new regimens of chemotherapeutic activities against skin cancer.
Collapse
Affiliation(s)
- Aeshah A Awaji
- Department of Biology, Faculty of Science, University College of Taymaa, University of Tabuk, Tabuk 71491, Saudi Arabia.
| | - Heba W Alhamdi
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | | | - Mohammad Y Alfaifi
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia; Tissue Culture and Cancer Biology Research Laboratory, King Khalid University, Abha 9004, Saudi Arabia.
| | - Ali A Shati
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia; Tissue Culture and Cancer Biology Research Laboratory, King Khalid University, Abha 9004, Saudi Arabia.
| | - Serag Eldin I Elbehairi
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia; Tissue Culture and Cancer Biology Research Laboratory, King Khalid University, Abha 9004, Saudi Arabia; Cell Culture Lab, Egyptian Organization for Biological Products and Vaccines (VACSERA Holding Company), 51 Wezaret El-Zeraa St., Agouza, Giza, Egypt.
| | - Nancy A-F Radwan
- Zoology Department, Faculty of Science, Arish University, 45511 El Arish, Egypt.
| | - Hani S Hafez
- Zoology Department, Faculty of Science, Suez University 43533, Suez, Egypt.
| | - Reda F M Elshaarawy
- Department of Chemistry, Faculty of Science, Suez University, 43533 Suez, Egypt; Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany.
| | - Mary Welson
- Zoology Department, Faculty of Science, Suez University 43533, Suez, Egypt
| |
Collapse
|
6
|
El Mahi Y, Nizami ZN, Wali AF, Al Neyadi A, Magramane M, Al Azzani M, Arafat K, Attoub S, Eid AH, Iratni R. Rhus coriaria induces autophagic and apoptotic cell death in pancreatic cancer cells. Front Pharmacol 2024; 15:1412565. [PMID: 39139643 PMCID: PMC11319293 DOI: 10.3389/fphar.2024.1412565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Background:Pancreatic cancer is a leading cause of cancer-related mortality worldwide with increasing global incidence. We previously reported the anticancer effect of Rhus coriaria ethanolic extract (RCE) in triple negative breast and colon cancer cells. Herein, we investigated the anticancer effect of RCE on human pancreatic cancer cells. Methods: Cell viability was measured using Cell Titer-Glo and staining of viable and dead cells based on differential permeability to two DNA binding dyes. Cell cycle distribution and annexin V staining was carried out in Muse cell analyzer. Protein level was determined by Western blot. Tumor growth was assessed by in ovo chick embryo chorioallantoic membrane assay. Results: We found that RCE significantly inhibited the viability and colony growth of pancreatic cancer cells (Panc-1, Mia-PaCa-2, S2-013, AsPC-1). The antiproliferative effects of RCE in pancreatic cancer cells (Panc-1 and Mia-PaCa-2) were mediated through induction of G1 cell cycle arrest, Beclin-1-independent autophagy, and apoptosis. RCE activated both the extrinsic and intrinsic pathways of apoptosis and regulated the Bax/Bcl-2 apoptotic switch. Mechanistically, we found that RCE inhibited the AKT/mTOR pathway, downstream of which, inactivation of the cell cycle regulator p70S6K and downregulation of the antiapoptotic protein survivin was observed. Additionally, we found that RCE-induced autophagy preceded apoptosis. Further, we confirmed the anticancer effect of RCE in a chick embryo xenograft model and found that RCE inhibited the growth of pancreatic cancer xenografts without affecting embryo survival. Conclusion: Collectively, our findings demonstrate that Rhus coriaria exerts potent anti-pancreatic cancer activity though cell cycle impairment, autophagy, and apoptosis, and is hence a promising source of anticancer phytochemicals.
Collapse
Affiliation(s)
- Yassine El Mahi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Zohra Nausheen Nizami
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Adil Farooq Wali
- Department of Pharmaceutical Chemistry, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Aysha Al Neyadi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohamed Magramane
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mazoun Al Azzani
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Kholoud Arafat
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Samir Attoub
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
7
|
Abu-Bakar A, Ismail M, Zulkifli MZI, Zaini NAS, Shukor NIA, Harun S, Inayat-Hussain SH. Mapping the influence of hydrocarbons mixture on molecular mechanisms, involved in breast and lung neoplasms: in silico toxicogenomic data-mining. Genes Environ 2024; 46:15. [PMID: 38982523 PMCID: PMC11232146 DOI: 10.1186/s41021-024-00310-y] [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: 11/20/2023] [Accepted: 06/07/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Exposure to chemical mixtures inherent in air pollution, has been shown to be associated with the risk of breast and lung cancers. However, studies on the molecular mechanisms of exposure to a mixture of these pollutants, such as hydrocarbons, in the development of breast and lung cancers are scarce. We utilized in silico toxicogenomic analysis to elucidate the molecular pathways linked to both cancers that are influenced by exposure to a mixture of selected hydrocarbons. The Comparative Toxicogenomics Database and Cytoscape software were used for data mining and visualization. RESULTS Twenty-five hydrocarbons, common in air pollution with carcinogenicity classification of 1 A/B or 2 (known/presumed or suspected human carcinogen), were divided into three groups: alkanes and alkenes, halogenated hydrocarbons, and polyaromatic hydrocarbons. The in silico data-mining revealed 87 and 44 genes commonly interacted with most of the investigated hydrocarbons are linked to breast and lung cancer, respectively. The dominant interactions among the common genes are co-expression, physical interaction, genetic interaction, co-localization, and interaction in shared protein domains. Among these genes, only 16 are common in the development of both cancers. Benzo(a)pyrene and tetrachlorodibenzodioxin interacted with all 16 genes. The molecular pathways potentially affected by the investigated hydrocarbons include aryl hydrocarbon receptor, chemical carcinogenesis, ferroptosis, fluid shear stress and atherosclerosis, interleukin 17 signaling pathway, lipid and atherosclerosis, NRF2 pathway, and oxidative stress response. CONCLUSIONS Within the inherent limitations of in silico toxicogenomics tools, we elucidated the molecular pathways associated with breast and lung cancer development potentially affected by hydrocarbons mixture. Our findings indicate adaptive responses to oxidative stress and inflammatory damages are instrumental in the development of both cancers. Additionally, ferroptosis-a non-apoptotic programmed cell death driven by lipid peroxidation and iron homeostasis-was identified as a new player in these responses. Finally, AHR potential involvement in modulating IL-8, a critical gene that mediates breast cancer invasion and metastasis to the lungs, was also highlighted. A deeper understanding of the interplay between genes associated with these pathways, and other survival signaling pathways identified in this study, will provide invaluable knowledge in assessing the risk of inhalation exposure to hydrocarbons mixture. The findings offer insights into future in vivo and in vitro laboratory investigations that focus on inhalation exposure to the hydrocarbons mixture.
Collapse
Affiliation(s)
- A'edah Abu-Bakar
- Product Stewardship and Toxicology, Environment, Social Performance & Product Stewardship (ESPPS), Group Health, Safety and Environment (GHSE), Petroliam Nasional Berhad (PETRONAS), Kuala Lumpur, 50088, Malaysia.
| | - Maihani Ismail
- Product Stewardship and Toxicology, Environment, Social Performance & Product Stewardship (ESPPS), Group Health, Safety and Environment (GHSE), Petroliam Nasional Berhad (PETRONAS), Kuala Lumpur, 50088, Malaysia.
| | - M Zaqrul Ieman Zulkifli
- Product Stewardship and Toxicology, Environment, Social Performance & Product Stewardship (ESPPS), Group Health, Safety and Environment (GHSE), Petroliam Nasional Berhad (PETRONAS), Kuala Lumpur, 50088, Malaysia
| | - Nur Aini Sofiyya Zaini
- Product Stewardship and Toxicology, Environment, Social Performance & Product Stewardship (ESPPS), Group Health, Safety and Environment (GHSE), Petroliam Nasional Berhad (PETRONAS), Kuala Lumpur, 50088, Malaysia
| | - Nur Izzah Abd Shukor
- Health, Safety and Environment (HSE), KLCC Urusharta, Kuala Lumpur, 50088, Malaysia
| | - Sarahani Harun
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, 43600 UKM, Malaysia
| | - Salmaan Hussain Inayat-Hussain
- ESPPS, GHSE, PETRONAS, Kuala Lumpur, 50088, Malaysia
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College St, New Haven, CT, 06250, USA
| |
Collapse
|
8
|
Athwal H, Kochiyanil A, Bhat V, Allan AL, Parsyan A. Centrosomes and associated proteins in pathogenesis and treatment of breast cancer. Front Oncol 2024; 14:1370565. [PMID: 38606093 PMCID: PMC11007099 DOI: 10.3389/fonc.2024.1370565] [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: 01/14/2024] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
Breast cancer is the most prevalent malignancy among women worldwide. Despite significant advances in treatment, it remains one of the leading causes of female mortality. The inability to effectively treat advanced and/or treatment-resistant breast cancer demonstrates the need to develop novel treatment strategies and targeted therapies. Centrosomes and their associated proteins have been shown to play key roles in the pathogenesis of breast cancer and thus represent promising targets for drug and biomarker development. Centrosomes are fundamental cellular structures in the mammalian cell that are responsible for error-free execution of cell division. Centrosome amplification and aberrant expression of its associated proteins such as Polo-like kinases (PLKs), Aurora kinases (AURKs) and Cyclin-dependent kinases (CDKs) have been observed in various cancers, including breast cancer. These aberrations in breast cancer are thought to cause improper chromosomal segregation during mitosis, leading to chromosomal instability and uncontrolled cell division, allowing cancer cells to acquire new genetic changes that result in evasion of cell death and the promotion of tumor formation. Various chemical compounds developed against PLKs and AURKs have shown meaningful antitumorigenic effects in breast cancer cells in vitro and in vivo. The mechanism of action of these inhibitors is likely related to exacerbation of numerical genomic instability, such as aneuploidy or polyploidy. Furthermore, growing evidence demonstrates enhanced antitumorigenic effects when inhibitors specific to centrosome-associated proteins are used in combination with either radiation or chemotherapy drugs in breast cancer. This review focuses on the current knowledge regarding the roles of centrosome and centrosome-associated proteins in breast cancer pathogenesis and their utility as novel targets for breast cancer treatment.
Collapse
Affiliation(s)
- Harjot Athwal
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Arpitha Kochiyanil
- Faculty of Science, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Vasudeva Bhat
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
| | - Alison L. Allan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Armen Parsyan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Division of General Surgery, Department of Surgery, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Surgery, St. Joseph’s Health Care London and London Health Sciences Centre, London, ON, Canada
| |
Collapse
|
9
|
Lambring CB, Fiadjoe H, Behera SK, Basha R. Docking and molecular dynamic simulations of Mithramycin-A and Tolfenamic acid against Sp1 and survivin. Process Biochem 2024; 137:207-216. [PMID: 38912413 PMCID: PMC11192519 DOI: 10.1016/j.procbio.2023.12.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Therapeutic targeting of Sp1 transcription factor and survivin, are studied in various cancers due to their consistent overexpression. These markers result in poorer cancer prognoses and their downregulation has been investigated as an effective treatment approach. Mithramycin-A and Tolfenamic acid are two drugs with innate anti-cancer properties and are suggested to be able to target Sp1 through GC/GT DNA binding interference, however in-depth binding and mechanistic studies are lacking. Through docking analysis, we investigated Mithramycin-A and Tolfenamic acid in terms of their specific binding interactions with Sp1 and survivin. Through further molecular dynamics simulations including Root Mean Square (RMS) Fluctuation and RMS Deviation, rGYr, and H-bond analysis, we identified critical residues involved in drug interactions with each protein in question. We show Mithramycin-A as the superior binding candidate to each protein and found that it exhibited stronger binding with Sp1, and then survivin. Subsequent molecular dynamics simulations followed the same trend as initial binding energy calculations and showed crucial amino acids involved in each Mithramycin-A-protein complex. Our findings warrant further investigation into Mithramycin-A and its specific interaction with Sp1 and their downstream targets giving a better understanding of Mithramycin-A and its potential as an effective cancer treatment.
Collapse
Affiliation(s)
| | - Hope Fiadjoe
- UNT Health Science Center at Fort Worth, Fort Worth, TX 76107, USA
| | | | - Riyaz Basha
- UNT Health Science Center at Fort Worth, Fort Worth, TX 76107, USA
| |
Collapse
|
10
|
Kondapuram SK, Ramachandran HK, Arya H, Coumar MS. Targeting survivin for cancer therapy: Strategies, small molecule inhibitors and vaccine based therapeutics in development. Life Sci 2023; 335:122260. [PMID: 37963509 DOI: 10.1016/j.lfs.2023.122260] [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: 10/04/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/16/2023]
Abstract
Survivin is a member of the family of inhibitors of apoptosis proteins (IAPs). It is involved in the normal mitotic process and acts as an anti-apoptotic molecule. While terminally differentiated normal tissues lack survivin, several human malignancies have significant protein levels. Resistance to chemotherapy and radiation in tumor cells is associated with survivin expression. Decreased tumor development, apoptosis, and increased sensitivity to chemotherapy and radiation are all effects of downregulating survivin expression or activity. As a prospective cancer treatment, small molecules targeting the transcription and translation of survivin and molecules that can directly bind with the survivin are being explored both in pre-clinical and clinics. Pre-clinical investigations have found and demonstrated the effectiveness of several small-molecule survivin inhibitors. Unfortunately, these inhibitors have also been shown to have off-target effects, which could limit their clinical utility. In addition to small molecules, several survivin peptide vaccines are currently under development. These vaccines are designed to elicit a cytotoxic T-cell response against survivin, which could lead to the destruction of tumor cells expressing survivin. Some survivin-based vaccines are advancing through Phase II clinical studies. Overall, survivin is a promising cancer drug target. However, challenges still need to be addressed before the survivin targeted therapies can be widely used in the clinics.
Collapse
Affiliation(s)
- Sree Karani Kondapuram
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry 605014, India
| | - Hema Kasthuri Ramachandran
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry 605014, India
| | - Hemant Arya
- Institute for Biochemistry and Pathobiochemistry, Department of Systems Biochemistry, Faculty of Medicine, Ruhr University Bochum, 44780 Bochum, Germany
| | - Mohane Selvaraj Coumar
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry 605014, India.
| |
Collapse
|
11
|
Cao Y, Tang H, Wang G, Li P, Song Z, Li W, Sun X, Zhong X, Yu Q, Zhu S, Zhu L. Targeting survivin with Tanshinone IIA inhibits tumor growth and overcomes chemoresistance in colorectal cancer. Cell Death Discov 2023; 9:351. [PMID: 37749082 PMCID: PMC10520088 DOI: 10.1038/s41420-023-01622-8] [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: 06/06/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 09/27/2023] Open
Abstract
The inhibitor of apoptosis protein survivin has a critical regulatory role in carcinogenesis and treatment tolerance in colorectal cancer (CRC). However, the targeted drugs for survivin protein are extremely limited. In the present research, we discovered that Tanshinone IIA (Tan IIA) played a dual regulatory role in inhibiting tumorigenesis and reversing 5-Fu tolerance via modulating the expression and phosphorylation of survivin in CRC cells. Mechanistically, Tan IIA suppressed the Akt/WEE1/CDK1 signaling pathway, which led to the downregulation of survivin Thr34 phosphorylation and destruction of the interaction between USP1 and survivin to promote survivin ubiquitination and degradation. Furthermore, Tan IIA significantly facilitated chemoresistant CRC cells to 5-Fu sensitivity. These results revealed that Tan IIA possessed a strong antitumor activity against CRC cells and could act as an up-and-coming agent for treating CRC and overcoming chemotherapy resistance.
Collapse
Affiliation(s)
- Yaoquan Cao
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Haibo Tang
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Guohui Wang
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Pengzhou Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Zhi Song
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Weizheng Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Xulong Sun
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Xiaoxiao Zhong
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Qianqian Yu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Shaihong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Liyong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
12
|
Quidute P, Quidute R, Perez MM, Pereira EC, da Veiga GL, Alves BDCA, Fonseca FLA. Survivin expression as a prognostic marker for breast cancer. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20230167. [PMID: 37729357 PMCID: PMC10508893 DOI: 10.1590/1806-9282.20230167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 07/03/2023] [Indexed: 09/22/2023]
Abstract
OBJECTIVE Due to the speed of development observed in breast cancer, several studies aimed at discovering new biomarkers have been carried out in order to arrive at an early diagnosis. As survivin plays a fundamental role in the evasion of apoptosis in tumor cells, the aim of this study was to verify the expression profile of the survivin gene in paraffin-embedded breast tumor samples and associate it with the clinical characteristics of the patients. METHODS This is a cross-sectional study, for which 100 tumor samples were obtained from cancer patients treated throughout the year 2019 at Instituto de Mama do Cariri (Juazeiro do Norte, in the state of Ceará). This study included women over 30 years old who had confirmed breast cancer through anatomopathological examination but excluded those with non-neoplastic breast comorbidities, other neoplasms, or chronic diseases. Survivin gene expression was assessed by quantitative polymerase chain reaction. RESULTS The expression of survivin is associated with the lack of expression of estrogen (p=0.027) and progesterone (p>0.0005) receptors. It means that survivin expression is higher in patients in which labeling was absent for estrogen receptor and progesterone receptor. CONCLUSION Our data reinforce that survivin expression is higher in estrogen receptor-patients, thus representing an additional prognostic tool.
Collapse
Affiliation(s)
- Patricia Quidute
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
| | - Ricardo Quidute
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
| | - Matheus Moreira Perez
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
| | | | - Glaucia Luciano da Veiga
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
| | | | - Fernando Luiz Affonso Fonseca
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
- Universidade Federal de São Paulo, Instituto de Ciências Farmacêuticas – Diadema (SP), Brazil
| |
Collapse
|
13
|
Wright S, Burkholz SR, Zelinsky C, Wittman C, Carback RT, Harris PE, Blankenberg T, Herst CV, Rubsamen RM. Survivin Expression in Luminal Breast Cancer and Adjacent Normal Tissue for Immuno-Oncology Applications. Int J Mol Sci 2023; 24:11827. [PMID: 37511584 PMCID: PMC10380623 DOI: 10.3390/ijms241411827] [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: 06/25/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Survivin (BIRC5) is a tumor-associated antigen (TAA) overexpressed in various tumors but present at low to undetectable levels in normal tissue. Survivin is known to have a high expression in breast cancer (e.g., Ductal Carcinoma in situ (DCIS) and triple negative breast cancer). Previous studies have not compared survivin expression levels in DCIS tumor samples to levels in adjacent, normal breast tissue from the same patient. To ensure the effective use of survivin as a target for T cell immunotherapy of breast cancer, it is essential to ascertain the varying levels of survivin expression between DCIS tumor tissue samples and the adjacent normal breast tissue taken from the same patient simultaneously. Next-generation sequencing of RNA (RNA-seq) in normal breast tissue and tumor breast tissue from five women presenting with DCIS for lumpectomy was used to identify sequence variation and expression levels of survivin. The identity of both tumor and adjacent normal tissue samples were corroborated by histopathology. Survivin was overexpressed in human breast tissue tumor samples relative to the corresponding adjacent human normal breast tissue. Wild-type survivin transcripts were the predominant species identified in all tumor tissue sequenced. This study demonstrates upregulated expression of wild type survivin in DCIS tumor tissue versus normal breast tissue taken from the same patient at the same time, and provides evidence that developing selective cytotoxic T lymphocyte (CTL) immunotherapy for DCIS targeting survivin warrants further study.
Collapse
Affiliation(s)
- Sharon Wright
- Saint Mary’s Regional Medical Center, Reno, NV 89503, USA; (S.W.); (C.Z.); (C.W.)
- Western Surgical Group, Reno, NV 89502, USA
| | - Scott R. Burkholz
- Flow Pharma Inc., Warrensville Heights, OH 44128, USA; (S.R.B.); (R.T.C.); (P.E.H.); (T.B.); (C.V.H.)
| | - Cathy Zelinsky
- Saint Mary’s Regional Medical Center, Reno, NV 89503, USA; (S.W.); (C.Z.); (C.W.)
| | - Connor Wittman
- Saint Mary’s Regional Medical Center, Reno, NV 89503, USA; (S.W.); (C.Z.); (C.W.)
| | - Richard T. Carback
- Flow Pharma Inc., Warrensville Heights, OH 44128, USA; (S.R.B.); (R.T.C.); (P.E.H.); (T.B.); (C.V.H.)
| | - Paul E. Harris
- Flow Pharma Inc., Warrensville Heights, OH 44128, USA; (S.R.B.); (R.T.C.); (P.E.H.); (T.B.); (C.V.H.)
| | - Tikoes Blankenberg
- Flow Pharma Inc., Warrensville Heights, OH 44128, USA; (S.R.B.); (R.T.C.); (P.E.H.); (T.B.); (C.V.H.)
- Shasta Pathology Associates, Redding, CA 96001, USA
| | - Charles V. Herst
- Flow Pharma Inc., Warrensville Heights, OH 44128, USA; (S.R.B.); (R.T.C.); (P.E.H.); (T.B.); (C.V.H.)
| | - Reid M. Rubsamen
- Saint Mary’s Regional Medical Center, Reno, NV 89503, USA; (S.W.); (C.Z.); (C.W.)
- Flow Pharma Inc., Warrensville Heights, OH 44128, USA; (S.R.B.); (R.T.C.); (P.E.H.); (T.B.); (C.V.H.)
- Cleveland Medical Center, University Hospitals, Cleveland, OH 44106, USA
- Case Western Reserve School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
14
|
Yu Z, Chao H, Xu F, Deng H, Deng L, Song Z, Zeng T. Identification of a prognostic biomarker predicting biochemical recurrence and construction of a novel nomogram for prostate cancer. Front Oncol 2023; 13:1115718. [PMID: 37077837 PMCID: PMC10106702 DOI: 10.3389/fonc.2023.1115718] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
BackgroundBiochemical recurrence (BCR) is common in prostate cancer (PCa), but its prediction is based predominantly on clinicopathological characteristics with low accuracy. We intend to identify a potential prognostic biomarker related to the BCR and construct a nomogram for improving the risk stratification of PCa patients.MethodsThe transcriptome and clinical data of PCa patients were obtained from TCGA and GEO databases. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were used to screen out differentially expressed genes (DEGs) related to the BCR of PCa. Cox regression analysis was further applied to screen out DEGs related to BCR-free survival (BFS). Time-dependent receiver operating curve (ROC) analysis and Kaplan–Meier (K-M) survival analysis were conducted to assess the prognostic value. Then, a prognostic nomogram was established and evaluated. The clinicopathological correlation analysis, GSEA analysis, and immune analysis were used to explore the biological and clinical significance of the biomarker. Finally, the qRT-PCR, western blotting, and immunohistochemistry (IHC) were conducted to validate the expression of the biomarker.ResultsBIRC5 was identified to be the potential prognostic biomarker. The clinical correlation analysis and K-M survival analysis found that the BIRC5 mRNA expression was positively associated with disease progression and negatively associated with the BFS rate. Time-dependent ROC curves verified its accurate prediction performance. The GSEA and immune analysis suggested that the BIRC5 was related to immunity. A nomogram with an accurate prediction for BFS of PCa patients was constructed. qRT-PCR, western blotting, and IHC results validated the expression level of BIRC5 in PCa cells and tissues.ConclusionOur study identified BIRC5 as a potential prognostic biomarker related to BCR of PCa and constructed an efficacy nomogram for predicting BFS to assist clinical decision-making.
Collapse
Affiliation(s)
- Zhaojun Yu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, JiangXi, China
- Medical Department, Nanchang University, Nanchang, JiangXi, China
| | - Haichao Chao
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, JiangXi, China
| | - Fanghua Xu
- Pathology Department, The People’s Hospital of Pingxiang, Pingxiang, JiangXi, China
| | - Huanhuan Deng
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, JiangXi, China
- Medical Department, Nanchang University, Nanchang, JiangXi, China
| | - Leihong Deng
- Department of Ultrasound, The First Affiliated Hospital of Nanchang University, Nanchang, JiangXi, China
| | - Zhen Song
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, JiangXi, China
| | - Tao Zeng
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, JiangXi, China
- *Correspondence: Tao Zeng,
| |
Collapse
|