1
|
Saloni, Sachan M, Rahul, Verma RS, Patel GK. SOXs: Master architects of development and versatile emulators of oncogenesis. Biochim Biophys Acta Rev Cancer 2025; 1880:189295. [PMID: 40058508 DOI: 10.1016/j.bbcan.2025.189295] [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: 10/02/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
Transcription factors regulate a variety of events and maintain cellular homeostasis. Several transcription factors involved in embryonic development, has been shown to be closely associated with carcinogenesis when deregulated. Sry-like high mobility group box (SOX) proteins are potential transcription factors which are evolutionarily conserved. They regulate downstream genes to determine cell fate, via various signaling pathways and cellular processes essential for tissue and organ development. Dysregulation of SOXs has been reported to promote or suppress tumorigenesis by modulating cellular reprogramming, growth, proliferation, angiogenesis, metastasis, apoptosis, immune modulation, lineage plasticity, maintenance of the stem cell pool, therapy resistance and cancer relapse. This review provides a crucial understanding of the molecular mechanism by which SOXs play multifaceted roles in embryonic development and carcinogenesis. It also highlights their potential in advancing therapeutic strategies aimed at targeting SOXs and their downstream effectors in various malignancies.
Collapse
Affiliation(s)
- Saloni
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Manisha Sachan
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Rahul
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, India
| | - Rama Shanker Verma
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India.
| | - Girijesh Kumar Patel
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India.
| |
Collapse
|
2
|
Karankar VS, Awasthi S, Srivastava N. Peptide-driven strategies against lung cancer. Life Sci 2025; 366-367:123453. [PMID: 39923837 DOI: 10.1016/j.lfs.2025.123453] [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: 11/11/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Lung cancer remains one of the most significant global health challenges, accounting for 18 % of all cancer-related deaths. While risk factors such as heavy metal exposure and cigarette smoking are well-known contributors, the limitations of conventional treatments including severe side effects and drug resistance highlight the urgent need for more targeted and safer therapeutic options. In this context, peptides have emerged as a novel, precise, and effective class of therapies for lung cancer treatment. They have shown promise in limiting lung cancer progression by targeting key molecular pathways involved in tumour growth. Anti-non-small cell lung cancer peptides that specifically target proteins such as EGFR, TP53, BRAF, MET, ROS1, and ALK have demonstrated potential in improving lung cancer outcomes. Additionally, anti-inflammatory and apoptosis-inducing peptides offer further therapeutic benefits. This review provides a comprehensive overview of the peptides currently in use or under investigation for the treatment of lung cancer, highlighting their mechanisms of action and therapeutic potential. As research continues to advance, peptides are poised to become a promising new therapeutic option in the fight against lung cancer.
Collapse
Affiliation(s)
- Vijayshree S Karankar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Saurabh Awasthi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India.
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India.
| |
Collapse
|
3
|
Yang Z, Chen M, Ge R, Zhou P, Pan W, Song J, Ma S, Chen S, Xu C, Zhou M, Mi W, Ni H, Chen H, Yao X, Dong X, Chen Y, Zhou J, Xuan C, Dong C, Yan H, Xie S. Identification of a non-inhibitory aptameric ligand to CRL2 ZYG11B E3 ligase for targeted protein degradation. Nat Commun 2025; 16:2494. [PMID: 40082426 PMCID: PMC11906892 DOI: 10.1038/s41467-025-57823-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 03/05/2025] [Indexed: 03/16/2025] Open
Abstract
As a crucial element of proteolysis targeting chimeras (PROTACs), the choice of E3 ubiquitin ligase significantly influences degradation efficacy and selectivity. However, the available arsenal of E3 ligases for PROTAC development remains underexplored, severely limiting the scope of targeted protein degradation. In this study, we identify a non-inhibitory aptamer targeting ZYG11B, a substrate receptor of the Cullin 2-RING ligase complex, as an E3 warhead for targeted protein degradation. This aptamer-based PROTAC platform, termed ZATAC, is facilely produced through bioorthogonal chemistry or self-assembly and shows promise in eliminating several undruggable target proteins, including nucleolin (NCL), SRY-box transcription factor 2 (SOX2), and mutant p53-R175H, underscoring its universality and versatility. To specifically deliver ZATACs into cancer cells, we further develop DNA three-way junction-based ZATACs (3WJ-ZATACs) by integrating an additional aptamer that selectively recognizes the protein overexpressed on the surface of cancer cells. The 3WJ-ZATACs demonstrate in vivo tumor-specific distribution and achieve dual-target degradation, thereby suppressing tumor growth without causing noticeable toxicity. In summary, ZATACs represent a general, modular, and straightforward platform for targeted protein degradation, offering insights into the potential of other untapped E3 ligases.
Collapse
Affiliation(s)
- Zhihao Yang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education); Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Biochemistry and Molecular Biology, Tianjin Medical University; Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Miao Chen
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| | - Ruixin Ge
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Ping Zhou
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Wei Pan
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jiayi Song
- Department of Ophthalmology, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuwen Ma
- Department of Ophthalmology, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin, China
| | - Song Chen
- Department of Ophthalmology, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin, China
| | - Chenyu Xu
- School of Medicine, Nankai University, Tianjin, China
| | - Mengyu Zhou
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Wenyi Mi
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hua Ni
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| | - He Chen
- Department of Medicinal Chemistry, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xue Yao
- Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopaedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xifeng Dong
- Department of Hematology, Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin Institute of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Chen
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jun Zhou
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| | - Chenghao Xuan
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education); Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Biochemistry and Molecular Biology, Tianjin Medical University; Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| | - Cheng Dong
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education); Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Biochemistry and Molecular Biology, Tianjin Medical University; Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| | - Hua Yan
- Department of Ophthalmology, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin, China.
| | - Songbo Xie
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education); Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Biochemistry and Molecular Biology, Tianjin Medical University; Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China.
- Department of Ophthalmology, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
4
|
Yang J, Zhu Y, Zhou Y, Zhang J, Wei Y, Liu Y, Zhang B, Xie J, An X, Qi X, Yue Y, Zhang L, Zhang X, Fu Z, Liu K. Potential biomarkers develop for predicting the prognosis of patients with esophageal squamous cell carcinoma after optimized chemoradiotherapy using serum metabolomics. BMC Cancer 2025; 25:438. [PMID: 40069698 PMCID: PMC11900641 DOI: 10.1186/s12885-025-13866-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC), the most common type of esophageal cancer, characterized by low five-year survival rate, and concurrent chemoradiotherapy (CCRT) has been proposed to treat ESCC, while potential biomarkers for prognostic monitoring after optimized CCRT remains unknown. METHODS Serum samples from 45 patients with ESCC were collected and categorized into three groups: Control (pre-CCRT), CCRT (during CCRT), and CCRT-1 M (one-month post-CCRT). The therapeutic effect was evaluated using CT imaging and established evaluation criteria. Untargeted metabolomic analysis was performed on the serum samples to identify differential metabolites caused by CCRT treatment, assessing their potential for prognostic monitoring. RESULTS CCRT had significant therapeutic efficacy in patients with ESCC, as indicated by CT imaging and RECIST 1.1 solid tumor evaluation criteria. Notably, several metabolic markers were identified through non-targeted metabolomic analysis, highlighting changes following CCRT treatment. These differential metabolites are involved in the dysregulation of phenylalanine, tyrosine, and tryptophan biosynthesis, as well as histidine, arginine, and proline metabolism, and glycine, serine, and threonine metabolism, suggesting a reduction in glucose metabolism in patients with ESCC after CCRT. Additionally, ROC analysis indicated that the AUC of these metabolites exceeded 0.661, underscoring their diagnostic value for assessing CCRT efficacy and their potential use in prognostic monitoring. Comparative metabolomic analysis identified L-phenylalanine and lysine as promising serum biomarkers for predicting therapeutic outcomes. CONCLUSIONS CCRT shows considerable therapeutic benefit in patients with ESCC, with observed reductions in glucose metabolism post-treatment. L-phenylalanine and lysine may serve as potential serum biomarkers to predict CCRT efficacy.
Collapse
Affiliation(s)
- Jie Yang
- Central Laboratory, Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu, 212300, P.R. China
| | - Yunyun Zhu
- Department of Radiotherapy, 900 Hospital of the Joint Logistics Team, (Dongfang Hospital, Xiamen University), Fuzhou, Fujian, 350025, P.R. China
| | - Yijian Zhou
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Jiaying Zhang
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Yuxuan Wei
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Yongpan Liu
- School of Life Science, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Bo Zhang
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Jialing Xie
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Xiaolu An
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Xianhua Qi
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Yuting Yue
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Lijia Zhang
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China
| | - Xiajun Zhang
- Central Laboratory, Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu, 212300, P.R. China.
| | - Zhichao Fu
- Department of Radiotherapy, 900 Hospital of the Joint Logistics Team, (Dongfang Hospital, Xiamen University), Fuzhou, Fujian, 350025, P.R. China.
| | - Kuancan Liu
- Central Laboratory, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, P.R. China.
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P.R. China.
| |
Collapse
|
5
|
Wang Z, Li R, Yang G, Wang Y. Cancer stem cell biomarkers and related signalling pathways. J Drug Target 2024; 32:33-44. [PMID: 38095181 DOI: 10.1080/1061186x.2023.2295222] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/10/2023] [Indexed: 12/20/2023]
Abstract
Cancer stem cells (CSCs) represent a distinct subset of neoplastic cells characterised by their heightened capacity for tumorigenesis. These cells are implicated in the facilitation of cancer metastasis, recurrence, and resistance to conventional therapeutic interventions. Extensive scientific research has been devoted to the identification of biomarkers and the elucidation of molecular mechanisms in order to improve targeted therapeutic approaches. Accurate identification of cancer stem cells based on biomarkers can provide a theoretical basis for drug combinations of malignant tumours. Targeted biomarker-based therapies also offer a silver lining for patients with advanced malignancies. This review aims comprehensively to consolidate the latest findings on CSCs biomarkers, targeted agents as well as biomarkers associated signalling pathways in well-established cancer types, thereby contributing to improved prognostic outcomes.
Collapse
Affiliation(s)
- Zhe Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Department of Infectious Disease, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Rui Li
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Guilin Yang
- Department of Infectious Disease, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Yijin Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
6
|
Cook E, Van de Vijver K, Parra-Herran C. Diagnosis of verruciform acanthotic vulvar intra-epithelial neoplasia (vaVIN) using CK17, SOX2 and GATA3 immunohistochemistry. Histopathology 2024; 84:1212-1223. [PMID: 38356340 DOI: 10.1111/his.15156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/16/2024]
Abstract
AIMS Verruciform acanthotic vulvar intra-epithelial neoplasia (vaVIN) is an HPV-independent, p53 wild-type lesion with distinct morphology and documented risk of recurrence and cancer progression. vaVIN is rare, and prospective distinction from non-neoplastic hyperplastic lesions can be difficult. CK17, SOX2 and GATA3 immunohistochemistry has emerging value in the diagnosis of HPV-independent lesions, particularly differentiated VIN. We aimed to test the combined value of these markers in the diagnosis of vaVIN versus its non-neoplastic differentials in the vulva. METHODS AND RESULTS CK17, SOX2 and GATA3 immunohistochemistry was evaluated on 16 vaVINs and 34 mimickers (verruciform xanthoma, lichen simplex chronicus, lichen sclerosus, psoriasis, pseudo-epitheliomatous hyperplasia). CK17 was scored as 3+ = full-thickness, 2+ = partial-thickness, 1+ = patchy, 0 = absent; SOX2 as 3+ = strong staining ≥ 10% cells, 2+ = moderate, 1 + =weak, 0 = staining in < 10% cells; and GATA3 as pattern 0 = loss in < 25% basal cells, 1 = loss in 25-75% basal cells, 2 = loss in > 75% basal cells. For analysis, results were recorded as positive (CK17 = 3+, SOX2 = 3+, GATA3 = patterns 1/2) or negative (CK17 = 2+/1+/0, SOX2 = 2+/1+/0, GATA3 = pattern 0). CK17, SOX2 and GATA3 positivity was documented in 81, 75 and 58% vaVINs, respectively, versus 32, 17 and 22% of non-neoplastic mimickers, respectively; ≥ 2 marker positivity conferred 83 sensitivity, 88 specificity and 86% accuracy in vaVIN diagnosis. Compared to vaVIN, SOX2 and GATA3 were differentially expressed in lichen sclerosus, lichen simplex chronicus and pseudo-epitheliomatous hyperplasia, whereas CK17 was differentially expressed in verruciform xanthoma and adjacent normal mucosa. CONCLUSIONS CK17, SOX2 and GATA3 can be useful in the diagnosis of vaVIN and its distinction from hyperplastic non-neoplastic vulvar lesions. Although CK17 has higher sensitivity, SOX2 and GATA3 are more specific, and the combination of all markers shows optimal diagnostic accuracy.
Collapse
Affiliation(s)
- Eleanor Cook
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | | | | |
Collapse
|
7
|
MacLean MR, Walker OL, Arun RP, Fernando W, Marcato P. Informed by Cancer Stem Cells of Solid Tumors: Advances in Treatments Targeting Tumor-Promoting Factors and Pathways. Int J Mol Sci 2024; 25:4102. [PMID: 38612911 PMCID: PMC11012648 DOI: 10.3390/ijms25074102] [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: 02/28/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer stem cells (CSCs) represent a subpopulation within tumors that promote cancer progression, metastasis, and recurrence due to their self-renewal capacity and resistance to conventional therapies. CSC-specific markers and signaling pathways highly active in CSCs have emerged as a promising strategy for improving patient outcomes. This review provides a comprehensive overview of the therapeutic targets associated with CSCs of solid tumors across various cancer types, including key molecular markers aldehyde dehydrogenases, CD44, epithelial cellular adhesion molecule, and CD133 and signaling pathways such as Wnt/β-catenin, Notch, and Sonic Hedgehog. We discuss a wide array of therapeutic modalities ranging from targeted antibodies, small molecule inhibitors, and near-infrared photoimmunotherapy to advanced genetic approaches like RNA interference, CRISPR/Cas9 technology, aptamers, antisense oligonucleotides, chimeric antigen receptor (CAR) T cells, CAR natural killer cells, bispecific T cell engagers, immunotoxins, drug-antibody conjugates, therapeutic peptides, and dendritic cell vaccines. This review spans developments from preclinical investigations to ongoing clinical trials, highlighting the innovative targeting strategies that have been informed by CSC-associated pathways and molecules to overcome therapeutic resistance. We aim to provide insights into the potential of these therapies to revolutionize cancer treatment, underscoring the critical need for a multi-faceted approach in the battle against cancer. This comprehensive analysis demonstrates how advances made in the CSC field have informed significant developments in novel targeted therapeutic approaches, with the ultimate goal of achieving more effective and durable responses in cancer patients.
Collapse
Affiliation(s)
- Maya R. MacLean
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Olivia L. Walker
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Raj Pranap Arun
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Wasundara Fernando
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
- Department of Biology, Acadia University, Wolfville, NS B4P 2R6, Canada
| | - Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Nova Scotia Health Authority, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
8
|
Jiang J, Wang Y, Sun M, Luo X, Zhang Z, Wang Y, Li S, Hu D, Zhang J, Wu Z, Chen X, Zhang B, Xu X, Wang S, Xu S, Huang W, Xia L. SOX on tumors, a comfort or a constraint? Cell Death Discov 2024; 10:67. [PMID: 38331879 PMCID: PMC10853543 DOI: 10.1038/s41420-024-01834-6] [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: 11/28/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
The sex-determining region Y (SRY)-related high-mobility group (HMG) box (SOX) family, composed of 20 transcription factors, is a conserved family with a highly homologous HMG domain. Due to their crucial role in determining cell fate, the dysregulation of SOX family members is closely associated with tumorigenesis, including tumor invasion, metastasis, proliferation, apoptosis, epithelial-mesenchymal transition, stemness and drug resistance. Despite considerable research to investigate the mechanisms and functions of the SOX family, confusion remains regarding aspects such as the role of the SOX family in tumor immune microenvironment (TIME) and contradictory impacts the SOX family exerts on tumors. This review summarizes the physiological function of the SOX family and their multiple roles in tumors, with a focus on the relationship between the SOX family and TIME, aiming to propose their potential role in cancer and promising methods for treatment.
Collapse
Affiliation(s)
- Junqing Jiang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yufei Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Mengyu Sun
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Xiangyuan Luo
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Zerui Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yijun Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Siwen Li
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Dian Hu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Jiaqian Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Zhangfan Wu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Xiaoping Chen
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases; Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, China
| | - Bixiang Zhang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases; Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, China
| | - Xiao Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Shuai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Westlake university school of medicine, Hangzhou, 310006, China
| | - Shengjun Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Wenjie Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases; Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, China.
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
9
|
Xiao Y, Zhang T, Zhang H. Recent advances in the peptide-based biosensor designs. Colloids Surf B Biointerfaces 2023; 231:113559. [PMID: 37738870 DOI: 10.1016/j.colsurfb.2023.113559] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/09/2023] [Accepted: 09/17/2023] [Indexed: 09/24/2023]
Abstract
Biosensors have rapidly emerged as a high-sensitivity and convenient detection method. Among various types of biosensors, optical and electrochemical are the most commonly used. Conventionally, antibodies have been employed to ensure specific interaction between the transmission material and analytes. However, there has been increasing recognition of peptides as a promising recognition element for biosensor development in recent years. The use of peptides as recognition elements provides high level of specificity, sensitivity, and stability for the detection process. The combination of peptide designs and optical or electrochemical detection methods has significantly improved biosensor efficacy. These advancements present opportunities for developing biosensors with diverse functions that can be used to lay a strong scientific foundation for the development of personalized medicine and various other fields. This paper reviews the recent advancements in the development and application of peptide-based optical and electrochemical biosensors, as well as their prospects as a sensor type.
Collapse
Affiliation(s)
- Yue Xiao
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, MOE Key Laboratory of Molecular Biophysics, Wuhan 430074, China
| | - Ting Zhang
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, MOE Key Laboratory of Molecular Biophysics, Wuhan 430074, China
| | - Houjin Zhang
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, MOE Key Laboratory of Molecular Biophysics, Wuhan 430074, China.
| |
Collapse
|
10
|
Chen Y, Zhang K, Zhang R, Wang Z, Yang L, Zhao T, Zhang S, Lin Y, Zhao H, Liu Y, Wei Y, Zhou Y, Zhang J, Ye X, Zhao J, Li X, Que J, Shi S, Liu K. Targeting the SOX2/CDP protein complex with a peptide suppresses the malignant progression of esophageal squamous cell carcinoma. Cell Death Discov 2023; 9:399. [PMID: 37891174 PMCID: PMC10611744 DOI: 10.1038/s41420-023-01693-7] [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: 05/31/2023] [Revised: 09/28/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Emerging evidence indicates that SOX2 is an oncogene for esophageal squamous cell carcinoma (ESCC). However, direct targeting of SOX2 is not feasible given that this transcription factor plays important roles in the maintenance of tissues such as the brain. Here, we identified CDP (Homeobox protein cut-like 1 or CASP) as a unique SOX2 binding partner enriched in ESCC with Duolink proximity ligation assay, bimolecular fluorescence complementation (BiFc) and immunoprecipitation. We then screened a peptide aptamer library using BiFc and immunoprecipitation and identified several peptide aptamers, including P58, that blocked the CDP/SOX2 interaction, leading to the inhibition of ESCC progress in vitro and in vivo. Upon administration, synthetic peptide P58, containing the YGRKKRRQRRR cell-penetrating peptide and the fluorophore TAMRA, also blocked the growth and metastasis of ESCC in both mice and zebrafish. Therefore, targeting the SOX2 binding partner CDP with peptide P58 offers an alternative avenue to treat ESCC with increased SOX2 levels.
Collapse
Affiliation(s)
- Yunyun Chen
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Kun Zhang
- Department of General Surgery, Fuzhou First General Hospital affiliated with Fujian Medical University, Fuzhou, Fujian, 350009, P. R. China
| | - Rui Zhang
- Department of Laboratory Medicine, The Second Hospital of Fuzhou, Fuzhou, Fujian, 350007, P. R. China
| | - Zhuo Wang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Liang Yang
- Westlake University, Hangzhou, Zhejiang, 310024, P. R. China
| | - Tingting Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Shihui Zhang
- Centre for Translational Stem Cell Biology, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, 999077, P. R. China
| | - Yong Lin
- Science and Technology Service Center, Fujian Health College, Fuzhou, Fujian, 350101, P. R. China
| | - Hongzhou Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Yongpan Liu
- School of Life Science, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Yuxuan Wei
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Yijian Zhou
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Jiaying Zhang
- School of Life Science, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Xianzong Ye
- Department of Pathology, 900 Hospital of the Joint Logistics Team (Dongfang Hospital, Xiamen University), Fuzhou, Fujian, 350025, P. R. China
| | - Jing Zhao
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Xinxin Li
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Jianwen Que
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Songlin Shi
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China.
| | - Kuancan Liu
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China.
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China.
- School of Life Science, Nanchang Normal University, Nanchang, Jiangxi, 330032, P. R. China.
| |
Collapse
|
11
|
Zeng Z, Fu M, Hu Y, Wei Y, Wei X, Luo M. Regulation and signaling pathways in cancer stem cells: implications for targeted therapy for cancer. Mol Cancer 2023; 22:172. [PMID: 37853437 PMCID: PMC10583419 DOI: 10.1186/s12943-023-01877-w] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/05/2023] [Indexed: 10/20/2023] Open
Abstract
Cancer stem cells (CSCs), initially identified in leukemia in 1994, constitute a distinct subset of tumor cells characterized by surface markers such as CD133, CD44, and ALDH. Their behavior is regulated through a complex interplay of networks, including transcriptional, post-transcriptional, epigenetic, tumor microenvironment (TME), and epithelial-mesenchymal transition (EMT) factors. Numerous signaling pathways were found to be involved in the regulatory network of CSCs. The maintenance of CSC characteristics plays a pivotal role in driving CSC-associated tumor metastasis and conferring resistance to therapy. Consequently, CSCs have emerged as promising targets in cancer treatment. To date, researchers have developed several anticancer agents tailored to specifically target CSCs, with some of these treatment strategies currently undergoing preclinical or clinical trials. In this review, we outline the origin and biological characteristics of CSCs, explore the regulatory networks governing CSCs, discuss the signaling pathways implicated in these networks, and investigate the influential factors contributing to therapy resistance in CSCs. Finally, we offer insights into preclinical and clinical agents designed to eliminate CSCs.
Collapse
Affiliation(s)
- Zhen Zeng
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Minyang Fu
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yuan Hu
- Department of Pediatric Nephrology Nursing, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Min Luo
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China.
| |
Collapse
|
12
|
Zhao H, Wei Y, Zhang J, Zhang K, Tian L, Liu Y, Zhang S, Zhou Y, Wang Z, Shi S, Fu Z, Fu J, Zhao J, Li X, Zhang L, Zhao L, Liu K. HPV16 infection promotes the malignant transformation of the esophagus and progression of esophageal squamous cell carcinoma. J Med Virol 2023; 95:e29132. [PMID: 37792307 DOI: 10.1002/jmv.29132] [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: 06/06/2023] [Revised: 07/27/2023] [Accepted: 09/05/2023] [Indexed: 10/05/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) may be correlated with HPV infection, and the mechanism underlying the ESCC formation induced by HPV16 infection remains elusive. Here, we overexpressed HPV16 E6 and E7 and coordinated the overexpression of these two genes in EPC2 and ESCC cells. We found that E7 and coordinated expression of E6 and E7 promoted the proliferation of EPC2 cells, and upregulation of shh was responsible for cell proliferation since the use of vismodegib led to the failure of organoid formation. Meanwhile, overexpression of E6 and E7 in ESCC cells promoted cell proliferation, migration, and invasion in vitro. Importantly, E6 and E7 coordinately increased the capability of tumor growth in nude mice, while vismodegib slowed the growth of tumors in NCG mice. Moreover, a series of genes and proteins changed in cell lines after overexpression of the E6 and E7 genes, the potential biological processes and pathways were systematically analyzed using a bioinformatics assay. Together, these findings suggest that the activation of the hedgehog pathway induced by HPV16 infection may initially transform basal cells in the esophagus and promote following malignant processes in ESCC cells. The application of hedgehog inhibitors may represent a therapeutic avenue for ESCC treatment.
Collapse
Affiliation(s)
- Hongzhou Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Yuxuan Wei
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Jiaying Zhang
- School of Life Science, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Kun Zhang
- Department of General Surgery, The First Hospital of Fuzhou, Fuzhou, Fujian, People's Republic of China
| | - Liming Tian
- Department of Gynecology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yongpan Liu
- School of Life Science, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Shihui Zhang
- Centre for Translational Stem Cell Biology, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Yijian Zhou
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Zhuo Wang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Songlin Shi
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Zhichao Fu
- Department of Radiotherapy, 900 Hospital of the Joint Logistics Team (Dongfang Hospital, Xiamen University), Fuzhou, Fujian, People's Republic of China
| | - Jianqian Fu
- Department of Medical Oncology, The Fifth Hospital of Xiamen, Xiamen, Fujian, People's Republic of China
| | - Jing Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Xinxin Li
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Lijia Zhang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Liran Zhao
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Kuancan Liu
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Life Science, Nanchang Normal University, Nanchang, Jiangxi, People's Republic of China
| |
Collapse
|
13
|
P U A, Raj G, John J, Mohan K M, John F, George J. Aptamers: Features, Synthesis and Applications. Chem Biodivers 2023; 20:e202301008. [PMID: 37709723 DOI: 10.1002/cbdv.202301008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/16/2023]
Abstract
Aptamers have become a topic of interest among the researchers and scientists since they not only possess all of the benefits of antibodies but also possess special qualities including heat stability, low cost, and limitless uses⋅ Here we give a review about the features, applications, and challenges of aptamers and also how they are beneficial over the antibodies for biomedical applications. Their unique features make aptamers a prominent tool in therapeutics, diagnostics, biosensors and targeted drug delivery. In conclusion, aptamers represent exciting materials for a variety of applications and can be modified to improve their properties and to extend their applications in biomedical field.
Collapse
Affiliation(s)
- Aiswarya P U
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Gopika Raj
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Jinju John
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Malavika Mohan K
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Franklin John
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Jinu George
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| |
Collapse
|
14
|
Song D, Ye Z, Chen F, Zhan L, Sun X. circFNDC3B promotes esophageal squamous cell carcinoma progression by targeting MYO5A via miR-370-3p/miR-136-5p. BMC Cancer 2023; 23:821. [PMID: 37667251 PMCID: PMC10476377 DOI: 10.1186/s12885-023-11314-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/18/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a prevalent malignant tumor worldwide. Circular RNA (circRNA) is of great value in tumorigenesis progression. However, the mechanism of circFNDC3B in ESCC remains to be clarified. METHODS Firstly, the circular characteristics of circFNDC3B were evaluated by Actinomycin D and RNase R measurements. The functions of circFNDC3B in ESCC cells were examined by CCK-8, EdU and flow cytometry. Subsequently, the molecular mechanism of circFNDC3B was explained using luciferase reporter gene detection. Finally, we constructed xenograft model to prove the role of circFNDC3B in vivo. RESULTS Our study revealed that circFNDC3B was more stable than its linear RNA and prominently upregulated in ESCC. Functional findings suggested that silencing of circFNDC3B reduced the proliferation and enhanced apoptosis of ESCC cells in vitro. Meanwhile, knockdown of circFNDC3B attenuated tumor progression in vivo. Next, miR-370-3p/miR-136-5p was discovered to bind circFNDC3B. miR-370-3p/miR-136-5p reversed the promotive effect on cell proliferation and the inhibitory effect on cell apoptosis of circFNDC3B. MYO5A was a downstream target of miR-370-3p/miR-136-5p. CircFNDC3B served as a sponge for miR-370-3p/miR-136-5p and alleviated the prohibitory effect of miR-370-3p/miR-136-5p on MYO5A, which accelerated ESCC progression. CONCLUSION circFNDC3B positively adjusted the MYO5A expression via spongy miR-370-3p/miR-136-5p, hence achieving the cancer-promoting effect on ESCC. circFNDC3B was a prospective diagnosis marker for ESCC.
Collapse
Affiliation(s)
- Dan Song
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, No.42, Baiziting, Nanjing, 210009, Jinagsu Province, China.
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| | - Ziqi Ye
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Fangyu Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Liangliang Zhan
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, No.42, Baiziting, Nanjing, 210009, Jinagsu Province, China
| | - Xinchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
15
|
Goh KW, Stephen A, Wu YS, Sim MS, Batumalaie K, Gopinath SC, Guad RM, Kumar A, Sekar M, Subramaniyan V, Fuloria NK, Fuloria S, Velaga A, Sarker MMR. Molecular Targets of Aptamers in Gastrointestinal Cancers: Cancer Detection, Therapeutic Applications, and Associated Mechanisms. J Cancer 2023; 14:2491-2516. [PMID: 37670975 PMCID: PMC10475355 DOI: 10.7150/jca.85260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/03/2023] [Indexed: 09/07/2023] Open
Abstract
Gastrointestinal (GI) cancers are among the most common cancers that impact the global population, with high mortality and low survival rates after breast and lung cancers. Identifying useful molecular targets in GI cancers are crucial for improving diagnosis, prognosis, and treatment outcomes, however, limited by poor targeting and drug delivery system. Aptamers are often utilized in the field of biomarkers identification, targeting, and as a drug/inhibitor delivery cargo. Their natural and chemically modifiable binding capability, high affinity, and specificity are favored over antibodies and potential early diagnostic imaging and drug delivery applications. Studies have demonstrated the use of different aptamers as drug delivery agents and early molecular diagnostic and detection probes for treating cancers. This review aims to first describe aptamers' generation, characteristics, and classifications, also providing insights into their recent applications in the diagnosis and medical imaging, prognosis, and anticancer drug delivery system of GI cancers. Besides, it mainly discussed the relevant molecular targets and associated molecular mechanisms involved, as well as their applications for potential treatments for GI cancers. In addition, the current applications of aptamers in a clinical setting to treat GI cancers are deciphered. In conclusion, aptamers are multifunctional molecules that could be effectively used as an anticancer agent or drug delivery system for treating GI cancers and deserve further investigations for clinical applications.
Collapse
Affiliation(s)
- Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, 71800 Nilai, Malaysia
| | - Annatasha Stephen
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Yuan Seng Wu
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Selangor 47500, Malaysia
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Selangor 47500, Malaysia
| | - Maw Shin Sim
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Kalaivani Batumalaie
- Department of Biomedical Sciences, Faculty of Health Sciences, Asia Metropolitan University, 81750 Johor Bahru, Malaysia
| | - Subash C.B. Gopinath
- Faculty of Chemical Engineering & Technology, Arau 02600, Institute of Nano Electronic Engineering, Kangar 01000, Micro System Technology, Centre of Excellence, Arau 02600, Pauh Campus, Universiti Malaysia Perlis (UniMAP), Perlis, Malaysia
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000 Kangar, Perlis, Malaysia
- Micro System Technology, Centre of Excellence (CoE), Universiti Malaysia Perlis (UniMAP), Pauh Campus, 02600 Arau, Perlis, Malaysia
- Department of Computer Science and Engineering, Faculty of Science and Information Technology, Daffodil International University, Daffodil Smart City, Birulia, Savar, Dhaka 1216, Bangladesh
| | - Rhanye Mac Guad
- Department of Biomedical Science and Therapeutics, Faculty of Medicine and Health Science, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | - Ashok Kumar
- Department of Internal Medicine, University of Kansas Medical Centre, Kansas City, Kansas 66103, United States
| | - Mahendran Sekar
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Malaysia
| | - Vetriselvan Subramaniyan
- Department of Pharmacology, Jeffrey Cheah School of Medicine and Health Sciences, MONASH University, Malaysia
- Department of Pharmacology, School of Medicine, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Selangor 42610, Malaysia
| | - Neeraj Kumar Fuloria
- Centre of Excellence for Biomaterials Engineering & Faculty of Pharmacy, AIMST University, Bedong 08100, Malaysia
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai 600077, India
| | - Shivkanya Fuloria
- Faculty of Pharmacy, AIMST University, Semeling, Bedong 08100, Malaysia
| | - Appalaraju Velaga
- Department of Medicinal Chemistry, Faculty of Pharmacy, MAHSA University, Selangor 42610, Malaysia
| | - Md. Moklesur Rahman Sarker
- Department of Pharmacy, State University of Bangladesh, 77 Satmasjid Road, Dhanmondi, Dhaka 1205, Bangladesh
- Health Med Science Research Network, 3/1, Block F, Lalmatia, Dhaka 1207, Bangladesh
| |
Collapse
|
16
|
Zhang X, Zhan S, Guan X, Zhang Y, Lu J, Yu Y, Jin Y, Yang Y, Chu P, Hong E, Yang H, Ren H, Geng D, Wang Y, Zhou P, Guo Y, Chang Y. TAF1D promotes proliferation by transcriptionally activating G2/M phase-related genes in MYCN-amplified neuroblastoma. Cancer Sci 2023. [PMID: 37094904 DOI: 10.1111/cas.15815] [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: 11/24/2022] [Revised: 03/18/2023] [Accepted: 04/05/2023] [Indexed: 04/26/2023] Open
Abstract
High-risk neuroblastoma (HR-NB) is an aggressive childhood cancer that responds poorly to currently available therapies and is associated with only about a 50% 5-year survival rate. MYCN amplification is a critical driver of these aggressive tumors, but so far there have not been any approved treatments to effectively treat HR-NB by targeting MYCN or its downstream effectors. Thus, the identification of novel molecular targets and therapeutic strategies to treat children diagnosed with HR-NB represents an urgent unmet medical need. Here, we conducted a targeted siRNA screening and identified TATA box-binding protein-associated factor RNA polymerase I subunit D, TAF1D, as a critical regulator of the cell cycle and proliferation in HR-NB cells. Analysis of three independent primary NB cohorts determined that high TAF1D expression correlated with MYCN-amplified, high-risk disease and poor clinical outcomes. TAF1D knockdown more robustly inhibited cell proliferation in MYCN-amplified NB cells compared with MYCN-non-amplified NB cells, as well as suppressed colony formation and inhibited tumor growth in a xenograft mouse model of MYCN-amplified NB. RNA-seq analysis revealed that TAF1D knockdown downregulates the expression of genes associated with the G2/M transition, including the master cell-cycle regulator, cell-cycle-dependent kinase 1 (CDK1), resulting in cell-cycle arrest at G2/M. Our findings demonstrate that TAF1D is a key oncogenic regulator of MYCN-amplified HR-NB and suggest that therapeutic targeting of TAF1D may be a viable strategy to treat HR-NB patients by blocking cell-cycle progression and the proliferation of tumor cells.
Collapse
Affiliation(s)
- Xuan Zhang
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Shijia Zhan
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xiaoxing Guan
- Department of Pathology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yanli Zhang
- Imaging Core Facility, Technology Center for Protein Science, Tsinghua University, Beijing, China
| | - Jie Lu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yongbo Yu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yaqiong Jin
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yeran Yang
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Ping Chu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Enyu Hong
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Hui Yang
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Huimin Ren
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Di Geng
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yadi Wang
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Pingping Zhou
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yan Chang
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
17
|
Zhang J, Wang Z, Zhao H, Wei Y, Zhou Y, Zhang S, Zhao J, Li X, Lin Y, Liu K. The roles of the SOX2 protein in the development of esophagus and esophageal squamous cell carcinoma, and pharmacological target for therapy. Biomed Pharmacother 2023; 163:114764. [PMID: 37100016 DOI: 10.1016/j.biopha.2023.114764] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/16/2023] [Accepted: 04/20/2023] [Indexed: 04/28/2023] Open
Abstract
SOX2 is a transcription factor belonging to the SOX gene family, whose activity has been associated with the maintenance of the stemness and self-renewal of embryonic stem cells (ESCs), as well as the induction of differentiated cells into induced pluripotent stem cells (iPSCs). Moreover, accumulating studies have shown that SOX2 is amplified in various cancers, notably in esophageal squamous cell carcinoma (ESCC). In addition, SOX2 expression is linked to multiple malignant processes, including proliferation, migration, invasion, and drug resistance. Taken together, targeting SOX2 might shed light on novel approaches for cancer therapy. In this review, we aim to summarize the current knowledge regarding SOX2 in the development of esophagus and ESCC. We also highlight several therapeutic strategies for targeting SOX2 in different cancer types, which can provide new tools to treat cancers possessing abnormal levels of SOX2 protein.
Collapse
Affiliation(s)
- Jiaying Zhang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Life Science, Xiamen University, Xiamen, Fujian 361102, China; Fujian Health College, Fuzhou, Fujian, 350101, China
| | - Zhuo Wang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; Fujian Health College, Fuzhou, Fujian, 350101, China
| | - Hongzhou Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; Fujian Health College, Fuzhou, Fujian, 350101, China
| | - Yuxuan Wei
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; Fujian Health College, Fuzhou, Fujian, 350101, China
| | - Yijian Zhou
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; Fujian Health College, Fuzhou, Fujian, 350101, China
| | - Shihui Zhang
- Centre for Translational Stem Cell Biology, School of Biomedical Sciences, The University of Hong Kong, Pokfulam 999077, Hong Kong, China; Fujian Health College, Fuzhou, Fujian, 350101, China
| | - Jing Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; Fujian Health College, Fuzhou, Fujian, 350101, China
| | - Xinxin Li
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; Fujian Health College, Fuzhou, Fujian, 350101, China
| | - Yong Lin
- Centre for Translational Stem Cell Biology, School of Biomedical Sciences, The University of Hong Kong, Pokfulam 999077, Hong Kong, China; Fujian Health College, Fuzhou, Fujian, 350101, China.
| | - Kuancan Liu
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; Fujian Health College, Fuzhou, Fujian, 350101, China.
| |
Collapse
|
18
|
Venugopal DC, Caleb CL, Kirupakaran NP, Shyamsundar V, Ravindran S, Yasasve M, Krishnamurthy A, Harikrishnan T, Sankarapandian S, Ramshankar V. Clinicopathological Significance of Cancer Stem Cell Markers (OCT-3/4 and SOX-2) in Oral Submucous Fibrosis and Oral Squamous Cell Carcinoma. Biomedicines 2023; 11:biomedicines11041040. [PMID: 37189658 DOI: 10.3390/biomedicines11041040] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Oral submucous fibrosis (OSMF) is highly prevalent in South East Asia with higher rates of malignant transformation in Indian subcontinent. Numerous biomarkers are now being studied to predict disease prognosis and detect malignant alterations at an early stage. Patients with clinically and biopsy-proven oral submucous fibrosis and oral squamous cell carcinoma were included in the study as the experimental group, while patients without a tobacco or betel nut habit who had their third molars surgically removed were included as the healthy control group. For the immunohistochemistry (IHC) investigation, 5-μm slices from formalin-fixed, paraffin-embedded tissue blocks (FFPE) were obtained. Fresh tissues (n = 45) from all three groups were collected and gene expression was studied using relative quantitation-based qPCR. The protein expression of octamer-binding transcription factor 3/4 (OCT 3/4) and sex-determining region Y-box 2 (SOX 2) was evaluated in the experimental group and compared with healthy controls. The IHC results showed a significant correlation with the expression of OCT 3/4 (p value = 0.000; χ2 = 20.244) and SOX 2 (p value = 0.006; χ2 = 10.101) among OSCC and OSMF patients in comparison to healthy controls. Both OCT 3/4 and SOX 2 showed overexpression of four-fold and three-fold in OSMF when compared to OSCC and healthy controls, respectively. This study shows the significant importance of cancer stem cell markers OCT 3/4 and SOX 2 to assess the disease prognosis in OSMF.
Collapse
|
19
|
Zhang S, Chen Y, Hu Q, Zhao T, Wang Z, Zhou Y, Wei Y, Zhao H, Wang J, Yang Y, Zhang J, Shi S, Zhang Y, Yang L, Fu Z, Liu K. SOX2 inhibits LLGL2 polarity protein in esophageal squamous cell carcinoma via miRNA-142-3p. Cancer Biol Ther 2022; 23:1-15. [PMID: 36131361 PMCID: PMC9519027 DOI: 10.1080/15384047.2022.2126248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 08/17/2022] [Accepted: 09/10/2022] [Indexed: 11/02/2022] Open
Abstract
ABBREVIATIONS CCK-8, Cell Counting Kit 8; Chip, Chromatin Immunoprecipitation; EC, Esophageal cancer; EMT, epithelial-to-mesenchymal transition; ESCC, Esophageal squamous cell carcinomas; LLGL2, lethal (2) giant larvae protein homolog 2; LLGL2ov, LLGL2 overexpression; MET, mesenchymal-epithelial transition; miRNAs, MicroRNAs; PRM-MS, Parallel reaction monitoring-Mass spectrometry; SD, Standard deviation; SOX, sex determining region Y (SRY)-like box; SOX2-Kd, SOX2-knockdwon; TUNEL, TdT-mediated dUTP Nick-End Labeling.
Collapse
Affiliation(s)
- Shihui Zhang
- Central Laboratory, Xiang’an Hospital of Xiamen University, Xiamen, China
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Yunyun Chen
- Central Laboratory, Xiang’an Hospital of Xiamen University, Xiamen, China
- School of Medicine, Xiamen University, Xiamen, China
| | - Qiong Hu
- School of Medicine, Xiamen University, Xiamen, China
- Department of Clinic Medical Laboratory, Zhoushan Hospital, Zhoushan, China
| | - Tingting Zhao
- Central Laboratory, Xiang’an Hospital of Xiamen University, Xiamen, China
- School of Medicine, Xiamen University, Xiamen, China
| | - Zhuo Wang
- Central Laboratory, Xiang’an Hospital of Xiamen University, Xiamen, China
- School of Medicine, Xiamen University, Xiamen, China
| | - Yijian Zhou
- Central Laboratory, Xiang’an Hospital of Xiamen University, Xiamen, China
- School of Medicine, Xiamen University, Xiamen, China
| | - Yuxuan Wei
- Central Laboratory, Xiang’an Hospital of Xiamen University, Xiamen, China
- School of Medicine, Xiamen University, Xiamen, China
| | - Hongzhou Zhao
- Central Laboratory, Xiang’an Hospital of Xiamen University, Xiamen, China
- School of Medicine, Xiamen University, Xiamen, China
| | - Junkai Wang
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Yaxin Yang
- Department of Biology, University of Rochester, Rochester, New York, USA
| | - Jiaying Zhang
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Songlin Shi
- School of Medicine, Xiamen University, Xiamen, China
| | - Yujun Zhang
- School of Medicine, Xiamen University, Xiamen, China
| | - Ling Yang
- School of Medicine, Xiamen University, Xiamen, China
| | - Zhichao Fu
- Department of radiotherapy, 900 Hospital of the Joint Logistics Team (Dongfang Hospital, Xiamen University), Fuzhou, China
| | - Kuancan Liu
- Central Laboratory, Xiang’an Hospital of Xiamen University, Xiamen, China
- School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
20
|
Xiang W, Peng Y, Zeng H, Yu C, Zhang Q, Liu B, Liu J, Hu X, Wei W, Deng M, Wang N, Liu X, Xie J, Hou W, Tang J, Long Z, Wang L, Liu J. Targeting treatment of bladder cancer using PTK7 aptamer-gemcitabine conjugate. Biomater Res 2022; 26:74. [PMID: 36471380 PMCID: PMC9721011 DOI: 10.1186/s40824-022-00328-9] [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: 08/28/2022] [Accepted: 11/22/2022] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND Gemcitabine (GEM) is one of the first-line chemotherapies for bladder cancer (BC), but the GEMs cannot recognize cancer cells and have a low long-term response rate and high recurrence rate with side effects during the treatment of BC. Targeted transport of GEMs to mediate cytotoxicity to tumor and avoid the systemic side effects remains a challenge in the treatment of BC. METHODS Based on a firstly confirmed biomarker in BC-protein tyrosine kinase 7 (PTK7), which is overexpressed on the cell membrane surface in BC cells, a novel targeting system protein tyrosine kinase 7 aptamer-Gemcitabine conjugate (PTK7-GEMs) was designed and synthesized using a specific PTK7 aptamer and GEM through auto-synthesis method to deliver GEM against BC. In addition, the antitumor effects and safety evaluation of PTK7-GEMs was assessed with a series of in vitro and in vivo assays. RESULTS PTK7-GEMs can specifically bind and enter to BC cells dependent on the expression levels of PTK7 and via the macropinocytosis pathway, which induced cytotoxicity after GEM cleavage from PTK7-GEMs respond to the intracellular phosphatase. Moreover, PTK7-GEMs showed stronger anti-tumor efficacy and excellent biosafety in three types of tumor xenograft mice models. CONCLUSION These results demonstrated that PTK7-GEMs is a successful targeted aptamer-drug conjugates strategy (APDCs) to treat BC, which will provide new directions for the precision treatment of BC in the field of biomarker-oriented tumor targeted therapy.
Collapse
Affiliation(s)
- Wei Xiang
- grid.431010.7Department of Urology, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013 China
| | - Yongbo Peng
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory for Pharmaceutical Metabolism Research, the Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, No.1, Yixueyuan Road, Chongqing, 400016 China
| | - Hongliang Zeng
- grid.431010.7Department of Urology, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013 China ,grid.489633.3Institute of Chinese Materia Medica, Hunan Academy of Chinese Medicine, No.8, Yuehua Road, Changsha, 410013 China
| | - Chunping Yu
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, Guangdong 510060 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, Guangdong 510060 China
| | - Qun Zhang
- grid.412615.50000 0004 1803 6239Department of Radiotherapy, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong 510080 China
| | - Biao Liu
- grid.431010.7Department of Urology, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013 China
| | - Jiahao Liu
- grid.431010.7Department of Urology, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013 China
| | - Xing Hu
- grid.431010.7Department of Urology, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013 China
| | - Wensu Wei
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, Guangdong 510060 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, Guangdong 510060 China
| | - Minhua Deng
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, Guangdong 510060 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, Guangdong 510060 China
| | - Ning Wang
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, Guangdong 510060 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, Guangdong 510060 China
| | - Xuewen Liu
- grid.431010.7Department of Onology, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013 China
| | - Jianfei Xie
- grid.431010.7Department of Nursing, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013 China
| | - Weibin Hou
- grid.431010.7Department of Urology, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013 China
| | - Jin Tang
- grid.431010.7Department of Urology, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013 China
| | - Zhi Long
- grid.431010.7Department of Urology, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013 China
| | - Long Wang
- grid.431010.7Department of Urology, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013 China
| | - Jianye Liu
- grid.431010.7Department of Urology, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, Hunan 410013 China
| |
Collapse
|
21
|
Ghasemii K, Darroudi M, Rahimmanesh I, Ghomi M, Hassanpour M, Sharifi E, Yousefiasl S, Ahmadi S, Zarrabi A, Borzacchiello A, Rabiee M, Paiva-Santos AC, Rabiee N. Advances in aptamer-based drug delivery vehicles for cancer therapy. BIOMATERIALS ADVANCES 2022; 140:213077. [PMID: 35952549 DOI: 10.1016/j.bioadv.2022.213077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 06/15/2023]
Abstract
Overall, aptamers are special classes of nucleic acid-based macromolecules that are beginning to investigate because of their capability of avidity binding to a specific target for clinical use. Taking advantage of target-specific medicine led to more effective therapeutic and limitation of side effects of drugs. Herein, we discuss several aptamers and their binding capability and capacity for selecting tumor biomarkers and usage of them as targeting ligands for the functionalization of nanomaterials. We review recent applications based on aptamers and several nanoparticles to rise efficacy and develop carrier systems such as graphene oxide, folic acid, gold, mesopores silica, and various polymers and copolymer, polyethylene glycol, cyclodextrin, chitosan. The nanocarriers have been characterized by particle size, zeta potential, aptamer conjugation, and drug encapsulation efficiency. Hydrodynamic diameter and Zeta potential can used in order to monitor aptamers' crosslinking, in-vitro drug release, intracellular delivery of nanocarriers, and cellular cytotoxicity assay. Also, they are studied for cellular uptake and internalization to types of cancer cell lines such as colorectal, breast, prostate, leukemia and etc. The results are investigated in in-vivo cytotoxicity assay and cell viability assay. Targeted cancer therapy seems a good and promising strategy to overcome the systemic toxicity of chemotherapy.
Collapse
Affiliation(s)
- Kousar Ghasemii
- Department of Organic Chemistry, Faculty of Chemistry, University of Mazandaran, Babolsar, Iran
| | - Mahdieh Darroudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Matineh Ghomi
- School of Chemistry, Damghan University, Damghan 36716-41167, Iran
| | - Mahnaz Hassanpour
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
| | - Esmaeel Sharifi
- Institute for Polymers, Composites and Biomaterials, National Research Council (IPCB-CNR), Naples 80125, Italy; Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, 6517838736 Hamadan, Iran
| | - Satar Yousefiasl
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, 6517838736 Hamadan, Iran
| | - Sepideh Ahmadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering & Natural Science, Istinye University, Sariyer 34396, Istanbul, Turkey
| | - Assunta Borzacchiello
- Institute for Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, 80125 Naples, Italy
| | - Mohammad Rabiee
- Biomaterial group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Navid Rabiee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea; School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia.
| |
Collapse
|
22
|
Wang Z, Wang J, Zhao H, Zhao T, Chen Y, Jiang M, Zhang S, Wei Y, Zhang J, Zhou Y, Shi S, Fu Z, Yang Y, Zhang Y, Yang L, Que J, Liu K. Targeting the SOX2/PARP1 complex to intervene in the growth of esophageal squamous cell carcinoma. Biomed Pharmacother 2022; 153:113309. [PMID: 35738180 DOI: 10.1016/j.biopha.2022.113309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022] Open
Abstract
Elevated SOX2 protein levels are closely correlated with the increased incidence of esophageal squamous cell carcinoma (ESCC). However, establishing effective target measures for ESCC treatments continue to be researched. It has been previously proposed that SOX2 represents a potential therapeutic target for ESCC. Here, we found that the enzyme Poly(ADP-Ribose) polymerase 1 (PARP1) enriched in ESCCs interact with SOX2. Inhibition of PARP1 with 3-aminobenzamide (3-ABA) or shRNA knockdown reduced the proliferation of ESCCs, accompanied by decreased protein levels of SOX2. RNA sequencing demonstrated that PARP1 inhibition affected multiple signaling pathways involved in cancer cell proliferation. Additionally, 3-ABA synergistically suppressed the growth of ESCC cells when combined with cisplatin, and metformin potentiated the suppressive effect of 3-ABA on ESCC cell growth. Together these findings suggest that targeting SOX2 binding partner PARP1 provides a possible avenue to treat patients with high levels of SOX2.
Collapse
Affiliation(s)
- Zhuo Wang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Junkai Wang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Hongzhou Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Tingting Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yunyun Chen
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Ming Jiang
- Department of Gastroenterology of The Children's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Shihui Zhang
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Yuxuan Wei
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Jiaying Zhang
- School of Life Science, Xiamen University, Xiamen, Fujian 361102, China
| | - Yijian Zhou
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Songlin Shi
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhichao Fu
- Department of radiotherapy, 900 Hospital of the Joint Logistics Team (Dongfang Hospital, Xiamen University), Fuzhou, Fujian 350025, China
| | - Yaxin Yang
- Department of Biology, University of Rochester, NY 14627, USA
| | - Yujun Zhang
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Ling Yang
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Jianwen Que
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.
| | - Kuancan Liu
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
23
|
Recent development of aptamer conjugated chitosan nanoparticles as cancer therapeutics. Int J Pharm 2022; 620:121751. [DOI: 10.1016/j.ijpharm.2022.121751] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/26/2022] [Accepted: 04/11/2022] [Indexed: 12/18/2022]
|
24
|
Pouremamali F, Vahedian V, Hassani N, Mirzaei S, Pouremamali A, Kazemzadeh H, Faridvand Y, Jafari-gharabaghlou D, Nouri M, Maroufi NF. The role of SOX family in cancer stem cell maintenance: With a focus on SOX2. Pathol Res Pract 2022; 231:153783. [DOI: 10.1016/j.prp.2022.153783] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/17/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023]
|
25
|
Enrichment of SOX2-Positive Cells in BRAF V600E Mutated and Recurrent Ameloblastoma. J Pers Med 2022; 12:jpm12010077. [PMID: 35055392 PMCID: PMC8780877 DOI: 10.3390/jpm12010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/01/2022] [Accepted: 01/05/2022] [Indexed: 11/17/2022] Open
Abstract
Ameloblastoma is the most common benign odontogenic neoplasm, but with an aggressive behavior and a high recurrence rate. Nowadays wide surgical resection is the current recommended treatment, which can cause further loss of function and esthetics. Recent studies point to the stem/progenitor cells as both initiators and propagators of the tumors. Elucidation of the cellular and molecular mechanisms underlying the tumor stem cells is of broad interest for understanding tumorigenesis and for developing effective targeted therapies. SRY related HMG box gene 2 (SOX2) is a transcription factor that plays important roles in development, stem cell renewal, and cancer formation. Few studies have revealed increased SOX2 expression in atypical ameloblastoma and ameloblastic carcinoma. For the development of personalized medicine for ameloblastoma, biomarkers that provide prognostic or predictive information regarding a tumor’s nature or its response to treatment are essential. Thus, in this study, we aimed to study if SOX2-positive cells exist in ameloblastomas and their correlation with the clinicopathologic parameters. Our data suggested BRAF(V600E) mutation might contribute to the expansion of SOX2-positive cells. The identification of BRAF(V600E) mutation and the amplification of SOX2-positive cells in ameloblastomas imply the possible benefit of applying BRAF and SOX2 inhibitors in recurrent and un-resectable ameloblastomas.
Collapse
|
26
|
Stevanovic M, Kovacevic-Grujicic N, Mojsin M, Milivojevic M, Drakulic D. SOX transcription factors and glioma stem cells: Choosing between stemness and differentiation. World J Stem Cells 2021; 13:1417-1445. [PMID: 34786152 PMCID: PMC8567447 DOI: 10.4252/wjsc.v13.i10.1417] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/15/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is the most common, most aggressive and deadliest brain tumor. Recently, remarkable progress has been made towards understanding the cellular and molecular biology of gliomas. GBM tumor initiation, progression and relapse as well as resistance to treatments are associated with glioma stem cells (GSCs). GSCs exhibit a high proliferation rate and self-renewal capacity and the ability to differentiate into diverse cell types, generating a range of distinct cell types within the tumor, leading to cellular heterogeneity. GBM tumors may contain different subsets of GSCs, and some of them may adopt a quiescent state that protects them against chemotherapy and radiotherapy. GSCs enriched in recurrent gliomas acquire more aggressive and therapy-resistant properties, making them more malignant, able to rapidly spread. The impact of SOX transcription factors (TFs) on brain tumors has been extensively studied in the last decade. Almost all SOX genes are expressed in GBM, and their expression levels are associated with patient prognosis and survival. Numerous SOX TFs are involved in the maintenance of the stemness of GSCs or play a role in the initiation of GSC differentiation. The fine-tuning of SOX gene expression levels controls the balance between cell stemness and differentiation. Therefore, innovative therapies targeting SOX TFs are emerging as promising tools for combatting GBM. Combatting GBM has been a demanding and challenging goal for decades. The current therapeutic strategies have not yet provided a cure for GBM and have only resulted in a slight improvement in patient survival. Novel approaches will require the fine adjustment of multimodal therapeutic strategies that simultaneously target numerous hallmarks of cancer cells to win the battle against GBM.
Collapse
Affiliation(s)
- Milena Stevanovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade 11042, Serbia
- Chair Biochemistry and Molecular Biology, Faculty of Biology, University of Belgrade, Belgrade 11158, Serbia
- Department of Chemical and Biological Sciences, Serbian Academy of Sciences and Arts, Belgrade 11000, Serbia.
| | - Natasa Kovacevic-Grujicic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade 11042, Serbia
| | - Marija Mojsin
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade 11042, Serbia
| | - Milena Milivojevic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade 11042, Serbia
| | - Danijela Drakulic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade 11042, Serbia
| |
Collapse
|
27
|
Wang Z, Zhao T, Zhang S, Wang J, Chen Y, Zhao H, Yang Y, Shi S, Chen Q, Liu K. The Wnt signaling pathway in tumorigenesis, pharmacological targets, and drug development for cancer therapy. Biomark Res 2021; 9:68. [PMID: 34488905 PMCID: PMC8422786 DOI: 10.1186/s40364-021-00323-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Wnt signaling was initially recognized to be vital for tissue development and homeostasis maintenance. Further studies revealed that this pathway is also important for tumorigenesis and progression. Abnormal expression of signaling components through gene mutation or epigenetic regulation is closely associated with tumor progression and poor prognosis in several tissues. Additionally, Wnt signaling also influences the tumor microenvironment and immune response. Some strategies and drugs have been proposed to target this pathway, such as blocking receptors/ligands, targeting intracellular molecules, beta-catenin/TCF4 complex and its downstream target genes, or tumor microenvironment and immune response. Here we discuss the roles of these components in Wnt signaling pathway in tumorigenesis and cancer progression, the underlying mechanisms that is responsible for the activation of Wnt signaling, and a series of drugs targeting the Wnt pathway provide multiple therapeutic values. Although some of these drugs exhibit exciting anti-cancer effect, clinical trials and systematic evaluation should be strictly performed along with multiple-omics technology.
Collapse
Affiliation(s)
- Zhuo Wang
- Central Laboratory, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361102, P. R. China.,School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Tingting Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361102, P. R. China.,School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Shihui Zhang
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, EH164UU, UK
| | - Junkai Wang
- Central Laboratory, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Yunyun Chen
- Central Laboratory, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361102, P. R. China.,School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Hongzhou Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361102, P. R. China.,School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Yaxin Yang
- Department of Biology, University of Rochester, Rochester, NY, 14627, USA
| | - Songlin Shi
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Qiang Chen
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, SAR, China
| | - Kuancan Liu
- Central Laboratory, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361102, P. R. China. .,School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China.
| |
Collapse
|
28
|
Huang Z, Chen H, Ye H, Chen Z, Jaffrezic-Renault N, Guo Z. An ultrasensitive aptamer-antibody sandwich cortisol sensor for the noninvasive monitoring of stress state. Biosens Bioelectron 2021; 190:113451. [PMID: 34171819 DOI: 10.1016/j.bios.2021.113451] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 11/19/2022]
Abstract
Cortisol is a major glucocorticoid that can affect physiological activities in the human body. Besides, it is also a biomarker that can reflect the stress state of the body. Therefore, in order to monitor stress states in a sensitive and non-invasive manner, an ultra-sensitive aptamer-antibody sandwich sensor modified with multi-walled carbon nanotubes, ordered mesoporous carbon CMK-3, and silver nanoparticles (MWCNTs/CMK-3/AgNPs) was proposed for non-invasive detection of cortisol in human saliva. The MWCNTs/CMK-3/AgNPs nanocomposite was fixed on the surface of the glassy carbon electrodes (GCEs) as the material for the first round of signal amplification, and secondary signal amplification was realized by conjugating cortisol antibodies with gold nanoparticles (AuNPs). Finally, the aptamer-antibody sandwich pattern was used to specifically recognize and bind cortisol. The concentration response range for this aptamer-antibody sandwich sensor is 0.1 pg/mL-10 ng/mL, and the limit of detection (LOD) is 0.09 pg/mL. So far, the LOD of this sensor has been relatively low, showing its good sensitivity, selectivity, stability, and reproducibility. Furthermore, it has been successfully applied to detect cortisol in saliva samples to compare the stress states of postgraduates and undergraduates.
Collapse
Affiliation(s)
- Ziyu Huang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, 430065, PR China; School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, PR China
| | - Hao Chen
- Department of Anaesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Huarong Ye
- China Resources and Wisco General Hospital, Wuhan, 430080, PR China
| | - Zixuan Chen
- Department of Clinical Medicine, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, PR China
| | - Nicole Jaffrezic-Renault
- University of Lyon, Institute of Analytical Sciences, UMR-CNRS 5280, 5, La Doua Street, Villeurbanne, 69100, France.
| | - Zhenzhong Guo
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, 430065, PR China.
| |
Collapse
|
29
|
Dasgupta S, Koljenović S, van den Bosch TPP, Swagemakers SMA, van der Hoeven NMA, van Marion R, van der Spek PJ, van Doorn HC, van Kemenade FJ, Ewing-Graham PC. Evaluation of Immunohistochemical Markers, CK17 and SOX2, as Adjuncts to p53 for the Diagnosis of Differentiated Vulvar Intraepithelial Neoplasia (dVIN). Pharmaceuticals (Basel) 2021; 14:ph14040324. [PMID: 33918187 PMCID: PMC8066509 DOI: 10.3390/ph14040324] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/26/2022] Open
Abstract
Histological diagnosis of differentiated vulvar intraepithelial neoplasia (dVIN), the precursor of human papillomavirus (HPV)-independent vulvar squamous cell carcinoma (VSCC), can be challenging, as features of dVIN may mimic those of non-dysplastic dermatoses. To aid the diagnosis, p53-immunohistochemistry (IHC) is commonly used, and mutant expression patterns are used to support a histological diagnosis of dVIN. However, a proportion of dVIN can show wild-type p53-expression, which is characteristic of non-dysplastic dermatoses. Furthermore, recent research has identified a novel precursor of HPV-independent VSCC—the p53-wild-type differentiated exophytic vulvar intraepithelial lesion (de-VIL). Currently, there are no established diagnostic IHC-markers for p53-wild-type dVIN or de-VIL. We evaluated IHC-markers, cytokeratin 17 (CK17), and SRY-box 2 (SOX2), as diagnostic adjuncts for dVIN. For this, IHC-expression of CK17, SOX2, and p53 was studied in dVIN (n = 56), de-VIL (n = 8), and non-dysplastic vulvar tissues (n = 46). For CK17 and SOX2, the percentage of cells showing expression, and the intensity and distribution of expression were recorded. We also performed next generation targeted sequencing (NGTS) on a subset of dVIN (n = 8) and de-VIL (n = 8). With p53-IHC, 74% of dVIN showed mutant patterns and 26% showed wild-type expression. Median percentage of cells expressing CK17 or SOX2 was significantly higher in dVIN (p53-mutant or p53-wild-type) and de-VIL than in non-dysplastic tissues (p < 0.01). Diffuse, moderate-to-strong, full epithelial expression of CK17 or SOX2 was highly specific for dVIN and de-VIL. With NGTS, TP53 mutations were detected in both dVIN and de-VIL. We infer that immunohistochemical markers CK17 and SOX2, when used along with p53, may help support the histological diagnosis of dVIN.
Collapse
Affiliation(s)
- Shatavisha Dasgupta
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
- Correspondence:
| | - Senada Koljenović
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
| | - Thierry P. P. van den Bosch
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
| | - Sigrid M. A. Swagemakers
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
- Department of Clinical Bioinformatics, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Nick M. A. van der Hoeven
- Department of Gynecology and Obstetrics, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands;
- Department of Gynecologic Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands;
| | - Ronald van Marion
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
| | - Peter J. van der Spek
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
- Department of Clinical Bioinformatics, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Helena C. van Doorn
- Department of Gynecologic Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands;
| | - Folkert J. van Kemenade
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
| | - Patricia C. Ewing-Graham
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.K.); (T.P.P.v.d.B.); (S.M.A.S.); (R.v.M.); (P.J.v.d.S.); (F.J.v.K.); (P.C.E.-G.)
| |
Collapse
|
30
|
Manibalan S, Thirukumaran K, Varshni M, Shobana A, Achary A. Report on biopharmaceutical profile of recent biotherapeutics and insilco docking studies on target bindings of known aptamer biotherapeutics. Biotechnol Genet Eng Rev 2021; 36:57-80. [PMID: 33393433 DOI: 10.1080/02648725.2020.1858395] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Accumulated Toxicity, disease recurrence and drug resistivity problems have been observed due to the synthetic and semisynthetic therapeutic practices, which alternatively led to focus on Bio-therapeutics production than xenobiotics. Quick plasma clearance and high potency are the reasons for trending research with huge pharma market of numerous Bio-therapeutics than ever before. Researchers proved that most of the nano and micro Bio-therapeutics have multiple beneficial therapeutic effects. We have analyzed the past, and present scenario of some notable clinically approved Bio-therapeutics to identify the future formulation needs with advanced techniques. Protein-related drugs are the foremost Bio-therapeutics such as antibodies, enzymes, and short, fragmented polypeptides show aggregation properties during storage, naked peptide moieties are resisted by the polar cell membrane, and also the antidrug antibodies were reported. Even though Nucleic acid nano-bodies are excellent target binders than proteins, they had only a few minutes of half-life. Maintaining homogeneousness upon storage of Bio-therapeutics is still a significant challenge in industrial-scale formulation. Notably, plant systems are identified as most useful cost-effective hosts to produce human enzymes than animal systems without any possible viral loads. Irrespective of numerous advancements in routes of administration and additives, subcutaneous is still a golden one to achieve better dynamics. Additionally, the interactions and effective bonds made by each class of well-known aptamer biotherapeutics which are considered as future drugs were studied.
Collapse
Affiliation(s)
- Subramaniyan Manibalan
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| | - Kandasamy Thirukumaran
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| | - Mathimaran Varshni
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| | - Ayyasamy Shobana
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| | - Anant Achary
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| |
Collapse
|
31
|
Jia J, Li H, Chu J, Sheng J, Wang C, Jia Z, Meng W, Yin H, Wan J, He F. LncRNA FAM83A-AS1 promotes ESCC progression by regulating miR-214/CDC25B axis. J Cancer 2021; 12:1200-1211. [PMID: 33442418 PMCID: PMC7797654 DOI: 10.7150/jca.54007] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Recent researches have pinpointed that long non-coding RNA (lncRNA) was tightly related to the carcinogenesis. However, the function of lncRNA in esophageal cell squamous carcinoma (ESCC) remains to be explored. In the current study, we assessed the expression pattern and the biological function of FAM83A-AS1 in ESCC. Methods: qRT-PCR was used to detect the expression of FAM83A-AS1, miR-214, and CDC25B expression in ESCC tissues and cell lines. CCK-8, transwell, apoptosis and cell cycle assays were performed to define the function of FAM83A-AS1 in ESCC cells. Furthermore, the regulation of miR-214 by FAM83A-AS1 was defined by qRT- PCR and rescue assays. In addition, the association between CDC25B, miR-214, CDC25B was confirmed by qRT-PCR. Results: Here, we discovered that FAM83A-AS1 was strongly expressed in ESCC tissues. FAM83A-AS1 abundance was associated with TNM stages and the differentiation grade of ESCC patients. The receiver operating characteristic curve (ROC) analysis indicated the high accuracy of FAM83A-AS1 in ESCC diagnosis. Functionally, inhibiting FAM83A-AS1 repressed cell proliferation, migration, and invasion in ESCC. In addition, we found that FAM83A-AS1 accelerated the cell cycle while inhibited cell apoptosis. Mechanistically, we found that FAM83A-AS1 regulated miR-214 expression, and there was a negative correlation between miR-214 and FAM83A-AS1 in ESCC. Rescue assay indicated that miR-214 could impair the suppressing effect of cell migration induced by FAM83A-AS1 depletion. Furthermore, CDC25B was a direct target of miR-214, and FAM83A-AS1 enhanced CDC25B expression while miR-214 positively CDC25B expression in ESCC. Conclusions: Collectively, we concluded that FAM83A-AS1 facilitated ESCC progression by regulating the miR-214/CDC25B axis. Our study showed FAM83A-AS1 may act as a promising target for ESCC diagnosis and therapy.
Collapse
Affiliation(s)
- Jinlin Jia
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Hongle Li
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jie Chu
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jinxiu Sheng
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Chang Wang
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Zimo Jia
- Department of Clinical Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Weiwei Meng
- Department of Blood Transfusion, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Huiqing Yin
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Junhu Wan
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Fucheng He
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
32
|
Cell-penetrating peptides in oncologic pharmacotherapy: A review. Pharmacol Res 2020; 162:105231. [PMID: 33027717 DOI: 10.1016/j.phrs.2020.105231] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/23/2020] [Accepted: 09/30/2020] [Indexed: 01/10/2023]
Abstract
Cancer is the second leading cause of death in the world and its treatment is extremely challenging, mainly due to its complexity. Cell-Penetrating Peptides (CPPs) are peptides that can transport into the cell a wide variety of biologically active conjugates (or cargoes), and are, therefore, promising in the treatment and in the diagnosis of several types of cancer. Some notable examples are TAT and Penetratin, capable of penetrating the central nervous system (CNS) and, therefore, acting in cancers of this system, such as Glioblastoma Multiforme (GBM). These above-mentioned peptides, conjugated with traditional chemotherapeutic such as Doxorubicin (DOX) and Paclitaxel (PTX), have also been shown to induce apoptosis of breast and liver cancer cells, as well as in lung cancer cells, respectively. In other cancers, such as esophageal cancer, the attachment of Magainin 2 (MG2) to Bombesin (MG2B), another CPP, led to pronounced anticancer effects. Other examples are CopA3, that selectively decreased the viability of gastric cancer cells, and the CPP p28. Furthermore, in preclinical tests, the anti-tumor efficacy of this peptide was evaluated on human breast cancer, prostate cancer, ovarian cancer, and melanoma cells in vitro, leading to high expression of p53 and promoting cell cycle arrest. Despite the numerous in vitro and in vivo studies with promising results, and the increasing number of clinical trials using CPPs, few treatments reach the expected clinical efficacy. Usually, their clinical application is limited by its poor aqueous solubility, immunogenicity issues and dose-limiting toxicity. This review describes the most recent advances and innovations in the use of CPPs in several types of cancer, highlighting their crucial importance for various purposes, from therapeutic to diagnosis. Further clinical trials with these peptides are warranted to examine its effects on various types of cancer.
Collapse
|
33
|
Functional characterization of SOX2 as an anticancer target. Signal Transduct Target Ther 2020; 5:135. [PMID: 32728033 PMCID: PMC7391717 DOI: 10.1038/s41392-020-00242-3] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/01/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
SOX2 is a well-characterized pluripotent factor that is essential for stem cell self-renewal, reprogramming, and homeostasis. The cellular levels of SOX2 are precisely regulated by a complicated network at the levels of transcription, post-transcription, and post-translation. In many types of human cancer, SOX2 is dysregulated due to gene amplification and protein overexpression. SOX2 overexpression is associated with poor survival of cancer patients. Mechanistically, SOX2 promotes proliferation, survival, invasion/metastasis, cancer stemness, and drug resistance. SOX2 is, therefore, an attractive anticancer target. However, little progress has been made in the efforts to discover SOX2 inhibitors, largely due to undruggable nature of SOX2 as a transcription factor. In this review, we first briefly introduced SOX2 as a transcription factor, its domain structure, normal physiological functions, and its involvement in human cancers. We next discussed its role in embryonic development and stem cell-renewal. We then mainly focused on three aspects of SOX2: (a) the regulatory mechanisms of SOX2, including how SOX2 level is regulated, and how SOX2 cross-talks with multiple signaling pathways to control growth and survival; (b) the role of SOX2 in tumorigenesis and drug resistance; and (c) current drug discovery efforts on targeting SOX2, and the future perspectives to discover specific SOX2 inhibitors for effective cancer therapy.
Collapse
|