1
|
Liu YY, Fu YF, Yang WY, Li Z, Lu Q, Su X, Shi J, Wu SQ, Liang D, He YT. DKK3 and SERPINB5 as novel serum biomarkers for gastric cancer: facilitating the development of risk prediction models for gastric cancer. Front Oncol 2025; 15:1536491. [PMID: 40231256 PMCID: PMC11994446 DOI: 10.3389/fonc.2025.1536491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/11/2025] [Indexed: 04/16/2025] Open
Abstract
The existing gastric cancer (GC) risk prediction models based on biomarkers are limited. This study aims to identify new promising biomarkers for GC to develop a risk prediction model for effective assessment, screening, and early diagnosis. This study was conducted utilizing a large combined cohort for upper gastrointestinal cancer that was established in Hebei Province, China. General macro risk factors, Helicobacter pylori (H.pylori) infection status, and protein biomarkers were collected through questionnaire surveys and laboratory tests. Novel GC biomarkers were explored using data-independent acquisition (DIA) proteomics and enzyme-linked immunosorbent assay (ELISA). Multiple machine learning algorithms were used to identify key predictors for the GC risk prediction model, which was validated with an independent external cohort from multiple hospitals. A total of 530 participants aged 40 to 74 were analyzed, with 104 ultimately diagnosed with GC. Significant biomarkers in GC patients were identified by DIA combined ELISA, including elevated Keratin 7 (KRT7) and Mammary fibrostatin (SERPINB5) (P<0.001) and decreased Dickkopf-associated protein 3 (DKK3) (P<0.001). Factors such as sex, age, smoking status, alcohol consumption, family history of GC, H. pylori infection, DKK3 and SERPINB5 were used to create a multidimensional risk prediction model for GC. This model achieved an area under the curve (AUC) of 0.938 (95% confidence interval: 0.913-0.962). The risk prediction model developed in this study shows high accuracy and practical utility, serving as an effective preliminary screening tool for identifying high-risk individuals for GC.
Collapse
Affiliation(s)
- Yan-Yu Liu
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yan-Fang Fu
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wan-Yu Yang
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zheng Li
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qian Lu
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xin Su
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jin Shi
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Si-Qi Wu
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Di Liang
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yu-Tong He
- School of Public Health, Hebei Medical University, Shijiazhuang, China
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
2
|
Sravani A, Chandrasekaran N, Thomas J, Mukherjee A. Formulation and characterization of cisplatin-loaded hydroxyl functionalized single-walled carbon nanotubes for targeting gastric cancer stem cells. Heliyon 2023; 9:e18798. [PMID: 37593603 PMCID: PMC10432176 DOI: 10.1016/j.heliyon.2023.e18798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 08/19/2023] Open
Abstract
Chemotherapy is the most commonly used therapeutic method for treating many malignancies including gastric cancer. Due to their non-specific and non-targeted drug delivery, it causes resistance leading to cancer progression, relapse, and metastasis of cancer. To overcome this problem we carried out a study aimed to develop a new cisplatin (Cisp) loaded hydroxyl functionalized single-walled carbon nanotube (OH-SWCNT) nanocarrier system to selectively eliminate gastric cancer stem cells. To our understanding, this is the first study of the non-covalent interaction of cisplatin loaded on the surface of hydroxyl-functionalized single-walled carbon nanotubes by ultrasonication. The physical and morphological characterization was carried out by UV-Vis, FTIR spectroscopy, and TEM. A sustained and controlled release of cisp from OH-SWCNT at all three pHs 3.5, 5.5, and 7.4 was observed. Gastric cancer stem cells were isolated from primary cells and were identified by using CD133+ and CD44+ specific markers. Cisplatin-loaded OH-SWCNT nanocarrier was capable of limiting the self-renewal capacity of both CD133+ and CD44+ populations and also decreasing the number of tumorspheres in gastric CSCs. The cell viability percent of AGS cells was 20% at 250 μg/ml concentration. The IC50 value was less than 50% mol/L at both 200 μg/ml and 250 μg/ml of cisplatin-loaded OH-SWCNT. Our findings suggest that cisplatin-loaded OH-SWCNT nanocarrier complexes could target gastric CSCs and also could provide a potential strategy for selectively targeting and efficiently eliminating gastric CSCs. This could be a promising approach to prevent gastric cancer recurrence and metastasis and also improve gastric cancer therapy.
Collapse
Affiliation(s)
- A.N.K.V. Sravani
- Centre for Nanobiotechnology, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
| | - Natarajan Chandrasekaran
- Centre for Nanobiotechnology, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
| | - John Thomas
- Centre for Nanobiotechnology, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
| | - Amitava Mukherjee
- Centre for Nanobiotechnology, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
| |
Collapse
|
3
|
An J, Hu X, Liu F. Current understanding of cancer stem cells: Immune evasion and targeted immunotherapy in gastrointestinal malignancies. Front Oncol 2023; 13:1114621. [PMID: 36910604 PMCID: PMC9996315 DOI: 10.3389/fonc.2023.1114621] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
As a relatively rare population of cancer cells existing in the tumor microenvironment, cancer stem cells (CSCs) possess properties of immune privilege to evade the attack of immune system, regulated by the microenvironment of CSCs, the so-called CSCs niche. The bidirectional interaction of CSCs with tumor microenvironment (TME) components favors an immunosuppressive shelter for CSCs' survival and maintenance. Gastrointestinal cancer stem cells (GCSCs) are broadly regarded to be intimately involved in tumor initiation, progression, metastasis and recurrence, with elevated tumor resistance to conventional therapies, which pose a major hindrance to the clinical efficacy for treated patients with gastrointestinal malignancies. Thus, a multitude of efforts have been made to combat and eradicate GCSCs within the tumor mass. Among diverse methods of targeting CSCs in gastrointestinal malignancies, immunotherapy represents a promising strategy. And the better understanding of GCSCs immunomodulation and immunoresistance mechanisms is beneficial to guide and design novel GCSCs-specific immunotherapies with enhanced immune response and clinical efficacy. In this review, we have gathered available and updated information to present an overview of the immunoevasion features harbored by cancer stem cells, and we focus on the description of immune escape strategies utilized by CSCs and microenvironmental regulations underlying CSCs immuno-suppression in the context of gastrointestinal malignancies. Importantly, this review offers deep insights into recent advances of CSC-targeting immunotherapeutic approaches in gastrointestinal cancers.
Collapse
Affiliation(s)
- Junyi An
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohua Hu
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Liu
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Agnarelli A, Vella V, Samuels M, Papanastasopoulos P, Giamas G. Incorporating Immunotherapy in the Management of Gastric Cancer: Molecular and Clinical Implications. Cancers (Basel) 2022; 14:cancers14184378. [PMID: 36139540 PMCID: PMC9496849 DOI: 10.3390/cancers14184378] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 01/30/2023] Open
Abstract
Simple Summary Gastric cancer is one of the most common malignant tumours worldwide, with the fifth and third highest morbidity and mortality, respectively, of all cancers. Survival is limited, as most of the patients are diagnosed at an advanced stage, and are not suitable for surgery with a curative intent. Chemotherapy has only modestly improved patients’ outcomes and is mainly given with a palliative intent. Immunotherapy has improved overall survival of patients with gastric cancer, and has thus become a new standard of care in clinic. In this review we discuss the strong molecular rationale for the administration of immunotherapy in this disease and analyse the clinical data supporting its use. Abstract Gastric cancer has a median survival of 11 months, and this poor prognosis has not improved over the last 30 years. Recent pre-clinical data suggest that there is high tumour-related neoantigen expression in gastric cancer cells, suggesting that a clinical strategy that enhances the host’s immune system against cancer cells may be a successful approach to improve clinical outcomes. Additionally, there has been an increasing amount of translational evidence highlighting the relevance of PD-L1 expression in gastric cancer cells, indicating that PD-1/PD-L1 inhibitors may be useful. Several molecular subgroups of gastric cancer have been identified to respond with excellent outcomes to immunotherapy, including microsatellite instable tumours, tumours bearing a high tumour mutational burden, and tumours related to a chronic EBV infection. In gastric cancer, immunotherapy has produced durable responses in chemo-refractory patients; however, most recently there has been a lot of enthusiasm as several large-scale clinical trials highlight the improved survival noted from the incorporation of immunotherapy in the first line setting for advanced gastric cancer. Our review aims to discuss current pre-clinical and clinical data supporting the innovative role of immunotherapy in gastric cancer.
Collapse
|
5
|
Ma ES, Wang ZX, Zhu MQ, Zhao J. Immune evasion mechanisms and therapeutic strategies in gastric cancer. World J Gastrointest Oncol 2022; 14:216-229. [PMID: 35116112 PMCID: PMC8790417 DOI: 10.4251/wjgo.v14.i1.216] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/22/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is a malignancy with a high incidence and mortality. The tumor immune microenvironment plays an important role in promoting cancer development and supports GC progression. Accumulating evidence shows that GC cells can exert versatile mechanisms to remodel the tumor immune microenvironment and induce immune evasion. In this review, we systematically summarize the intricate crosstalk between GC cells and immune cells, including tumor-associated macrophages, neutrophils, myeloid-derived suppressor cells, natural killer cells, effector T cells, regulatory T cells, and B cells. We focus on how GC cells alter these immune cells to create an immunosuppressive microenvironment that protects GC cells from immune attack. We conclude by compiling the latest progression of immune checkpoint inhibitor-based immunotherapies, both alone and in combination with conventional therapies. Anti-cytotoxic T-lymphocyte-associated protein 4 and anti-programmed cell death protein 1/programmed death-ligand 1 therapy alone does not provide substantial clinical benefit for GC treatment. However, the combination of immune checkpoint inhibitors with chemotherapy or targeted therapy has promising survival advantages in refractory and advanced GC patients. This review provides a comprehensive understanding of the immune evasion mechanisms of GC, and highlights promising immunotherapeutic strategies.
Collapse
Affiliation(s)
- En-Si Ma
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institute of Organ Transplantation, Fudan University, Shanghai 200040, China
| | - Zheng-Xin Wang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institute of Organ Transplantation, Fudan University, Shanghai 200040, China
| | - Meng-Qi Zhu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jing Zhao
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| |
Collapse
|
6
|
Ni T, Wang H, Zhan D, Tao L, Lv M, Wang W, Chu Z, Zhou Z, Sunagawa M, Liu Y. CD133+/CD166+ human gastric adenocarcinoma cells present the properties of neoplastic stem cells and emerge more malignant features. Life Sci 2021; 269:119021. [PMID: 33450261 DOI: 10.1016/j.lfs.2021.119021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/31/2020] [Accepted: 01/06/2021] [Indexed: 12/28/2022]
Abstract
AIMS The recurrence and metastasis of gastric cancer has always been an important factor affecting the prognosis of gastric cancer. Cancer stem cells can promote the recurrence and growth of gastric cancer. The identification and isolation of gastric cancer stem cells contribute to the origin, progress and treatment strategy of gastric cancer. The aim of this study was to identify and isolate gastric cancer stem cells, and provide targets for the treatment of gastric cancer. METHODS Magnetic-activated cell sorting was used to isolate CD133+/CD166+ cell populations from human gastric adenocarcinoma cell lines (BGC-823 and SGC-7901). Sphere formation, cell proliferation, resistance to chemotherapy, colony formation, migration invasion and tumorigenicity in vivo of these cell populations were evaluated. Moreover, RT-qPCR and Western blot were used to investigate the expression level of the stem cell markers Nanog, Sox2, Oct-4, and c-Myc. RESULTS CD133+/CD166+ cell subpopulations presented more malignant features than CD133-/CD166-, CD133-/CD166+, CD133+/CD166- cell populations and parental cells. Moreover, the mRNA and protein expression level of Oct-4 and c-Myc were higher in CD133+/CD166+ cells than in parental cells or other cell populations. CONCLUSION The CD133+/CD166+ populations of human gastric cancer cell lines BGC-823 and SGC-7901 have cancer stem cell characteristics.
Collapse
Affiliation(s)
- Tengyang Ni
- Institute of Translational Medicine, Medical College, Yangzhou University, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China
| | - Haibo Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China
| | - Dongmei Zhan
- Institute of Translational Medicine, Medical College, Yangzhou University, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China
| | - Li Tao
- Department of Pharmacy, College of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Mengying Lv
- Department of Pharmacy, College of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Weimin Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China
| | - Zewen Chu
- Institute of Translational Medicine, Medical College, Yangzhou University, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China
| | - Zhen Zhou
- Institute of Translational Medicine, Medical College, Yangzhou University, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China
| | - Masataka Sunagawa
- Department of Physiology, School of Medicine, Showa University, Tokyo 142, Japan
| | - Yanqing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China.
| |
Collapse
|
7
|
Nie XC, He F, Lan C, Niu JM, Xia P. Combined Serum DKK3 and Circulating CD133 Cells as Prognostic Biomarkers for Ovarian Cancer Patients. Onco Targets Ther 2021; 14:427-434. [PMID: 33488097 PMCID: PMC7814242 DOI: 10.2147/ott.s288191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/21/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction Ovarian cancer (OV) can seriously endanger women’s physical and mental health. Serum DKK3 has been used for the diagnosis and prognosis of patients with ovarian cancer. However, the specificity of antibodies may lead to errors in the detection of plasma protein. Methods Circulating CD133+ cells from blood samples were separated by magnetic microbeads. Serum DKK3 levels were determined by ELISA. The roles of DKK3 in OV cells were analyzed in vitro. Results In this study, we found that the CD133+ subpopulation in circulating tumor cells can indicate the overall survival rate of OV patients. Serum DKK3 levels were negatively correlated with the number of circulating CD133+ cells in OV patients. In addition, we confirmed the inhibitory effect of recombinant human DKK3 (rhDKK3) on OV cells via reversal of the epithelial–mesenchymal transition (EMT) process. Conclusion Both serum DKK3 levels and circulating CD133+ tumor cells can be used as prognostic markers for patients with ovarian cancer.
Collapse
Affiliation(s)
- Xiao-Cui Nie
- Department of Gynaecology, Shenyang Women's and Children's Hospital, Shenyang, Liaoning, People's Republic of China
| | - Fang He
- Department of Gynaecology, Shenyang Women's and Children's Hospital, Shenyang, Liaoning, People's Republic of China
| | - Chong Lan
- Department of Gynaecology, Shenyang Women's and Children's Hospital, Shenyang, Liaoning, People's Republic of China
| | - Ju-Min Niu
- Department of Gynaecology, Shenyang Women's and Children's Hospital, Shenyang, Liaoning, People's Republic of China
| | - Pu Xia
- Biological Anthropology Institute, Liaoning Medical University, Jinzhou, Liaoning, People's Republic of China
| |
Collapse
|
8
|
Ferretti E, Carlomagno S, Pesce S, Muccio L, Obino V, Greppi M, Solari A, Setti C, Marcenaro E, Della Chiesa M, Sivori S. Role of the Main Non HLA-Specific Activating NK Receptors in Pancreatic, Colorectal and Gastric Tumors Surveillance. Cancers (Basel) 2020; 12:E3705. [PMID: 33321719 PMCID: PMC7763095 DOI: 10.3390/cancers12123705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022] Open
Abstract
Human NK cells can control tumor growth and metastatic spread thanks to their powerful cytolytic activity which relies on the expression of an array of activating receptors. Natural cytotoxicity receptors (NCRs) NKG2D and DNAM-1 are those non-HLA-specific activating NK receptors that are mainly involved in sensing tumor transformation by the recognition of different ligands, often stress-induced molecules, on the surface of cancer cells. Tumors display several mechanisms aimed at dampening/evading NK-mediated responses, a relevant fraction of which is based on the downregulation of the expression of activating receptors and/or their ligands. In this review, we summarize the role of the main non-HLA-specific activating NK receptors, NCRs, NKG2D and DNAM-1, in controlling tumor growth and metastatic spread in solid malignancies affecting the gastrointestinal tract with high incidence in the world population, i.e., pancreatic ductal adenocarcinoma (PDAC), colorectal cancer (CRC), and gastric cancer (GC), also describing the phenotypic and functional alterations induced on NK cells by their tumor microenvironment.
Collapse
Affiliation(s)
- Elisa Ferretti
- Centro di Eccellenza per la Ricerca Biomedica, University of Genoa, 16132 Genoa, Italy;
| | - Simona Carlomagno
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Silvia Pesce
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Letizia Muccio
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Valentina Obino
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Marco Greppi
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Agnese Solari
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Chiara Setti
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Emanuela Marcenaro
- Dipartimento di Medicina Sperimentale (DIMES) and Centro di Eccellenza per la Ricerca Biomedica, University of Genoa, 16132 Genoa, Italy;
| | - Mariella Della Chiesa
- Dipartimento di Medicina Sperimentale (DIMES) and Centro di Eccellenza per la Ricerca Biomedica, University of Genoa, 16132 Genoa, Italy;
| | - Simona Sivori
- Dipartimento di Medicina Sperimentale (DIMES) and Centro di Eccellenza per la Ricerca Biomedica, University of Genoa, 16132 Genoa, Italy;
| |
Collapse
|
9
|
Shen F, Gan XX, Deng XY, Feng JH, Cai WS, Shen L, Xiao HQ, Xu B. MicroRNA-3690 promotes cell proliferation and cell cycle progression by altering DKK3 expression in human thyroid cancer. Oncol Lett 2020; 20:223. [PMID: 32968445 PMCID: PMC7500009 DOI: 10.3892/ol.2020.12086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 07/20/2020] [Indexed: 12/20/2022] Open
Abstract
An increasing amount of evidence has demonstrated the importance of microRNAs (miRNAs/miRs) in the tumorigenesis of malignant types of cancer, and data retrieved from The Cancer Genome Atlas database revealed that miR-3690 was upregulated in thyroid cancer (TC). The present study focused on the biological function and mechanism of miR-3690 in TC, demonstrating that miR-3690 expression was significantly elevated in TC cells and clinical tissues. Functional studies indicated that miR-3690 acted as an oncogene in TC by promoting cell proliferation, colony formation and cell cycle progression in association with the increased expression of cyclin E and c-myc. Mechanistically, prediction software indicated that Dickkopf-related protein 3 (DKK3) was a target of miR-3690, which was confirmed by the results of luciferase reporter assays and western blotting. DKK3 silencing abrogated the functions of miR-3690-in on TC cell proliferation. Collectively, the findings of the present study demonstrated that miR-3690 promoted TC cell proliferation and indicated miR-3690 as a potential biomarker and therapeutic target for TC.
Collapse
Affiliation(s)
- Fei Shen
- Department of Thyroid Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China.,Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| | - Xiao-Xiong Gan
- Department of Thyroid Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China.,Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| | - Xing-Yan Deng
- Department of Thyroid Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China.,Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| | - Jian-Hua Feng
- Department of Thyroid Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China.,Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| | - Wen-Song Cai
- Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| | - Liang Shen
- Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| | - Huan-Qing Xiao
- Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| | - Bo Xu
- Department of Thyroid Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China.,Department of Thyroid Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| |
Collapse
|
10
|
Giuli MV, Hanieh PN, Giuliani E, Rinaldi F, Marianecci C, Screpanti I, Checquolo S, Carafa M. Current Trends in ATRA Delivery for Cancer Therapy. Pharmaceutics 2020; 12:E707. [PMID: 32731612 PMCID: PMC7465813 DOI: 10.3390/pharmaceutics12080707] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/23/2020] [Accepted: 07/25/2020] [Indexed: 12/12/2022] Open
Abstract
All-Trans Retinoic Acid (ATRA) is the most active metabolite of vitamin A. It is critically involved in the regulation of multiple processes, such as cell differentiation and apoptosis, by activating specific genomic pathways or by influencing key signaling proteins. Furthermore, mounting evidence highlights the anti-tumor activity of this compound. Notably, oral administration of ATRA is the first choice treatment in Acute Promyelocytic Leukemia (APL) in adults and NeuroBlastoma (NB) in children. Regrettably, the promising results obtained for these diseases have not been translated yet into the clinics for solid tumors. This is mainly due to ATRA-resistance developed by cancer cells and to ineffective delivery and targeting. This up-to-date review deals with recent studies on different ATRA-loaded Drug Delivery Systems (DDSs) development and application on several tumor models. Moreover, patents, pre-clinical, and clinical studies are also reviewed. To sum up, the main aim of this in-depth review is to provide a detailed overview of the several attempts which have been made in the recent years to ameliorate ATRA delivery and targeting in cancer.
Collapse
Affiliation(s)
- Maria Valeria Giuli
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy; (M.V.G.); (E.G.); (I.S.)
| | - Patrizia Nadia Hanieh
- Department of Drug Chemistry and Technology, Sapienza University of Rome, 00185 Rome, Italy; (P.N.H.); (F.R.); (C.M.); (M.C.)
| | - Eugenia Giuliani
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy; (M.V.G.); (E.G.); (I.S.)
| | - Federica Rinaldi
- Department of Drug Chemistry and Technology, Sapienza University of Rome, 00185 Rome, Italy; (P.N.H.); (F.R.); (C.M.); (M.C.)
| | - Carlotta Marianecci
- Department of Drug Chemistry and Technology, Sapienza University of Rome, 00185 Rome, Italy; (P.N.H.); (F.R.); (C.M.); (M.C.)
| | - Isabella Screpanti
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy; (M.V.G.); (E.G.); (I.S.)
| | - Saula Checquolo
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University of Rome, 04100 Latina, Italy
| | - Maria Carafa
- Department of Drug Chemistry and Technology, Sapienza University of Rome, 00185 Rome, Italy; (P.N.H.); (F.R.); (C.M.); (M.C.)
| |
Collapse
|
11
|
A1CF-promoted colony formation and proliferation of RCC depends on DKK1-MEK/ERK signal axis. Gene 2019; 730:144299. [PMID: 31881249 DOI: 10.1016/j.gene.2019.144299] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 12/14/2019] [Accepted: 12/16/2019] [Indexed: 12/23/2022]
Abstract
The function and mechanism of RNA editing proteins have been extensively studied, but its association with cellular processes and signaling pathways remained unaddressed. Here, we explored the function of RNA editing complementary protein- Apobec-1 Complementation Factor (A1CF) in the proliferation and colony formation of renal cell carcinoma (RCC) cells. Decreased A1CF expression inhibits the proliferation and colony formation of 786-O cells; and further signaling pathway screening demonstrated that A1CF increases ERK activation and DKK1 expression. Moreover, knockdown of DKK1 has similar phenotypes with A1CF deficiency in 786-O cells on cell proliferation and colony formation and ERK activation. Decreasing of DKK1 expression reduces the phosphorylation of ERK1/2 and MEK1/2 increased by A1CF overexpression; further, inhibiting of the phosphorylation of MEK1/2 by U0126 also decreases the ERK activation upregulated by A1CF overexpression. Deficiency of DKK1 or U0126 treatment suppresses the cell proliferation promoted by A1CF overexpression in 786-O cells; furthermore, U0126 treatment inhibits DKK1-increased cell proliferation in 786-O cells. Our results reveal that DKK1 mediates A1CF to activate ERK in promotion renal carcinoma cell proliferation and colony formation. For the important function of ERK signaling pathway in tumor metastasis and key position of DKK1 in Wnt signaling pathway, we associate RNA editing protein-A1CF with multiple cellular processes and signaling pathways through DKK1, and the key node of A1CF-DKK1-MEK/ERK axis is a potential targeting site for RCC therapy.
Collapse
|
12
|
Yang Y, Xu W, Zheng Z, Cao Z. LINC00459 sponging miR-218 to elevate DKK3 inhibits proliferation and invasion in melanoma. Sci Rep 2019; 9:19139. [PMID: 31844121 PMCID: PMC6914790 DOI: 10.1038/s41598-019-55701-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/27/2019] [Indexed: 12/22/2022] Open
Abstract
The lncRNA biomarkers in melanoma remain to be further explored. The lncRNAs with different expression levels in melanoma tissue were identified by microarray analysis. To investigate the biological functions of target lncRNA, several in-vivo and in-vitro studies were performed. Potential mechanisms of competitive endogenous RNAs (ceRNAs) were predicted by using bioinformatics analysis and explored by western blot assay, fluorescence in situ hybridization assay, real-time quantitative PCR (RT-qPCR) array, RNA pull-down analysis, AGO2-RIP assay, and dual-luciferase reporter assay. The results demonstrated decreased LINC00459 in melanoma cell lines and tissues. According to the in-vitro and in-vivo experiments, up-regulated LINC00459 had inhibitory effect on cell proliferation and invasion. Bioinformatics analyses suggested that miR-218 could be a direct target of LINC00459. In addition, miR-218 was proved to be able to directly target the dickkopf-related protein 3 (DKK3) gene. In conclusion, our analysis suggested that the LINC00459 could sponge miR-218 and increase the expression of DKK3 gene, thus inhibiting the invasion and proliferation of melanoma cells, which indicated that the LINC00459 could be an effective biomarker for melanoma and its potential as the therapeutic target.
Collapse
Affiliation(s)
- Yuhua Yang
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wenxian Xu
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhuojun Zheng
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Zhihai Cao
- Department of Emergency, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| |
Collapse
|
13
|
The promotion of nanoparticle delivery to two populations of gastric cancer stem cells by CD133 and CD44 antibodies. Biomed Pharmacother 2019; 115:108857. [DOI: 10.1016/j.biopha.2019.108857] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 12/16/2022] Open
|
14
|
Du Y, Wei Y. Therapeutic Potential of Natural Killer Cells in Gastric Cancer. Front Immunol 2019; 9:3095. [PMID: 30719024 PMCID: PMC6348255 DOI: 10.3389/fimmu.2018.03095] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 12/13/2018] [Indexed: 12/19/2022] Open
Abstract
Gastric cancer (GC) is one of the most common cancers, with a high incidence of cancer death. Despite various therapeutic approaches, the cures and prognosis of advanced GC remain poor. Natural killer (NK) cells, which are known as important lymphocytes in innate immunity, play vital roles in suppressing GC initiation, progression, and metastases. A wide range of clinical settings shows that increasing the number of NK cells or improving NK cell antitumor activity is promising in GC patients. NK cell adoptive therapy (especially expanded NK cells) is a safe and well-tolerated method, which can enhance NK cell cytotoxicity against GC. Meanwhile, cytokines, immunomodulatory drugs, immune checkpoint blockades, antibodies, vaccines, and gene therapy have been found to directly or indirectly activate NK cells to improve their killing activity toward GC. In this review, we summarize recent advancements in the relationship between NK cells and GC and point out all the innovative strategies that can enhance NK cells' function to inhibit the growth of GC.
Collapse
Affiliation(s)
- Yu Du
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yongchang Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|