1
|
Chen B, Liu J. Advances in ovarian tumor stem cells and therapy. Cell Biochem Biophys 2024; 82:1871-1892. [PMID: 38955927 DOI: 10.1007/s12013-024-01385-8] [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] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
Ovarian cancer is considered the most lethal among all gynecological malignancies due to its early metastatic dissemination, extensive spread, and malignant ascites. The current standard of care for advanced ovarian cancer involves a combination of cytoreductive surgery and chemotherapy utilizing platinum-based and taxane-based agents. Although initial treatment yields clinical remission in 70-80% of patients, the majority eventually develop treatment resistance and tumor recurrence. A growing body of evidence indicates the existence of cancer stem cells within diverse solid tumors, including ovarian cancer, which function as a subpopulation to propel tumor growth and disease advancement by means of drug resistance, recurrence, and metastasis. The presence of ovarian cancer stem cells is widely considered to be a significant contributor to the unfavorable clinical outcomes observed in patients with ovarian cancer, as they play a crucial role in mediating chemotherapy resistance, recurrence, and metastasis. Ovarian cancer stem cells possess the capacity to reassemble within the entirety of the tumor following conventional treatment, thereby instigating the recurrence of ovarian cancer and inducing resistance to treatment. Consequently, the creation of therapeutic approaches aimed at eliminating ovarian cancer stem cells holds great potential for the management of ovarian cancer. These cells are regarded as one of the most auspicious targets and mechanisms for the treatment of ovarian cancer. There is a pressing need for a comprehensive comprehension of the fundamental mechanisms of ovarian cancer's recurrence, metastasis, and drug resistance, alongside the development of effective strategies to overcome chemoresistance, metastasis, and recurrence. The implementation of cancer stem cell therapies may potentially augment the tumor cells' sensitivity to existing chemotherapy protocols, thereby mitigating the risks of tumor metastasis and recurrence, and ultimately improving the survival rates of ovarian cancer patients.
Collapse
Affiliation(s)
- Biqing Chen
- Harbin Medical University, Harbin, Heilongjiang, China.
| | - Jiaqi Liu
- Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
2
|
Wang Y, Chen X, Yang Y. CircRNA-regulated glucose metabolism in ovarian cancer: an emerging landscape for therapeutic intervention. Clin Transl Oncol 2024; 26:584-596. [PMID: 37578652 DOI: 10.1007/s12094-023-03285-4] [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: 05/11/2023] [Accepted: 07/11/2023] [Indexed: 08/15/2023]
Abstract
Ovarian cancer (OC) has the highest mortality rate among female reproductive system tumours, with limited efficacy of traditional treatments and 5-year survival rates that rarely exceed 40%. Circular RNA (circRNA) is a stable endogenous circular RNA that typically regulates protein expression by binding to downstream miRNA. It has been demonstrated that circRNAs play an important role in the proliferation, migration, and glucose metabolism (such as the Warburg effect) of OC and can regulate the expression of glucose metabolism-related proteins such as GLUT1 and HK2, promoting anaerobic glycolysis of cancer cells, increasing glucose uptake and ATP production, and affecting energy supply and biosynthetic substances to support tumour growth and invasion. This review summarises the formation and characteristics of circRNAs and focuses on their role in regulating glucose metabolism in OC cells and their potential therapeutic value, providing insights for identifying new therapeutic targets.
Collapse
Affiliation(s)
- Yaolong Wang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Gynecological Oncology of Gansu Province, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Xi Chen
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Gynecological Oncology of Gansu Province, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yongxiu Yang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
- Key Laboratory of Gynecological Oncology of Gansu Province, Lanzhou, Gansu, China.
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
3
|
RAGE Inhibitors for Targeted Therapy of Cancer: A Comprehensive Review. Int J Mol Sci 2022; 24:ijms24010266. [PMID: 36613714 PMCID: PMC9820344 DOI: 10.3390/ijms24010266] [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: 09/17/2022] [Revised: 10/28/2022] [Accepted: 12/16/2022] [Indexed: 12/28/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE) is a member of the immunoglobulin family that is overexpressed in several cancers. RAGE is highly expressed in the lung, and its expression increases proportionally at the site of inflammation. This receptor can bind a variety of ligands, including advanced glycation end products, high mobility group box 1, S100 proteins, adhesion molecules, complement components, advanced lipoxidation end products, lipopolysaccharides, and other molecules that mediate cellular responses related to acute and chronic inflammation. RAGE serves as an important node for the initiation and stimulation of cell stress and growth signaling mechanisms that promote carcinogenesis, tumor propagation, and metastatic potential. In this review, we discuss different aspects of RAGE and its prominent ligands implicated in cancer pathogenesis and describe current findings that provide insights into the significant role played by RAGE in cancer. Cancer development can be hindered by inhibiting the interaction of RAGE with its ligands, and this could provide an effective strategy for cancer treatment.
Collapse
|
4
|
Yang H, Guo Y, Zhang Y, Wang D, Zhang G, Hou J, Yang J. Circ_MUC16 attenuates the effects of Propofol to promote the aggressive behaviors of ovarian cancer by mediating the miR-1182/S100B signaling pathway. BMC Anesthesiol 2021; 21:297. [PMID: 34837947 PMCID: PMC8626908 DOI: 10.1186/s12871-021-01517-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 11/16/2021] [Indexed: 01/20/2023] Open
Abstract
Background Propofol is commonly used for anesthesia during surgery and has been demonstrated to inhibit cancer development, which is shown to be associated with deregulation of non-coding RNAs (ncRNAs). The objective of this study was to explore the role of circular RNA mucin 16 (circ_MUC16) in Propofol-mediated inhibition of ovarian cancer. Methods The expression of circ_MUC16, microRNA-1182 (miR-1182) and S100 calcium-binding protein B (S100B) mRNA was measured by quantitative real-time polymerase chain reaction (qPCR). The expression of S100B protein was checked by western blot. Cell proliferation was assessed by 3-(4, 5-di methyl thiazol-2-yl)-2, 5-di phenyl tetrazolium bromide (MTT) assay and colony formation assay. Glycolysis metabolism was assessed by glucose consumption, lactate production and ATP level. Cell migration and cell invasion were assessed by transwell assay. Cell migration was also assessed by wound healing assay. Animal study was conducted in nude mice to determine the role of circ_MUC16 in vivo. The relationship between miR-1182 and circ_MUC16 or S100B was validated by dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. Results Propofol inhibited ovarian cancer cell proliferation, glycolysis metabolism, migration and invasion, which were partly recovered by circ_MUC16 overexpression. Circ_MUC16 was downregulated in Propofol-treated ovarian cancer cells. Besides, circ_MUC16 knockdown enhanced the effects of Propofol to further inhibit tumor growth in vivo. MiR-1182 was a target of circ_MUC16, and circ_MUC16 knockdown-inhibited cell proliferation, glycolysis metabolism, migration and invasion were partly restored by miR-1182 inhibition. In addition, S100B was a target of miR-1182, and miR-1182-suppressed cell proliferation, glycolysis metabolism, migration and invasion were partly restored by S100B overexpression. Conclusion Circ_MUC16 overexpression alleviated the effects of Propofol to promote the aggressive behaviors of ovarian cancer by targeting the miR-1182/S100B network. Supplementary Information The online version contains supplementary material available at 10.1186/s12871-021-01517-0.
Collapse
Affiliation(s)
- Hao Yang
- Department of Anesthesiology, The Second People's Hospital of Kunming, Kunming College, No. 871, Longquan Road, Kunming, 650200, Yunnan, China
| | - Yunrui Guo
- Department of Anesthesiology, The Second People's Hospital of Kunming, Kunming College, No. 871, Longquan Road, Kunming, 650200, Yunnan, China
| | - Yecai Zhang
- Department of Anesthesiology, The Second People's Hospital of Kunming, Kunming College, No. 871, Longquan Road, Kunming, 650200, Yunnan, China
| | - Decai Wang
- Department of Anesthesiology, The Second People's Hospital of Kunming, Kunming College, No. 871, Longquan Road, Kunming, 650200, Yunnan, China
| | - Guoyun Zhang
- Department of Anesthesiology, The Second People's Hospital of Kunming, Kunming College, No. 871, Longquan Road, Kunming, 650200, Yunnan, China
| | - Jiali Hou
- Department of Anesthesiology, The Second People's Hospital of Kunming, Kunming College, No. 871, Longquan Road, Kunming, 650200, Yunnan, China
| | - Jianming Yang
- Department of Anesthesiology, The Second People's Hospital of Kunming, Kunming College, No. 871, Longquan Road, Kunming, 650200, Yunnan, China.
| |
Collapse
|
5
|
A Novel S100 Family-Based Signature Associated with Prognosis and Immune Microenvironment in Glioma. JOURNAL OF ONCOLOGY 2021; 2021:3586589. [PMID: 34712325 PMCID: PMC8548170 DOI: 10.1155/2021/3586589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/13/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022]
Abstract
Background Glioma is the most common central nervous system (CNS) cancer with a short survival period and a poor prognosis. The S100 family gene, comprising 25 members, relates to diverse biological processes of human malignancies. Nonetheless, the significance of S100 genes in predicting the prognosis of glioma remains largely unclear. We aimed to build an S100 family-based signature for glioma prognosis. Methods We downloaded 665 and 313 glioma patients, respectively, from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) database with RNAseq data and clinical information. This study established a prognostic signature based on the S100 family genes through multivariate COX and LASSO regression. The Kaplan-Meier curve was plotted to compare overall survival (OS) among groups, whereas Receiver Operating Characteristic (ROC) analysis was performed to evaluate model accuracy. A representative gene S100B was further verified by in vitro experiments. Results An S100 family-based signature comprising 5 genes was constructed to predict the glioma that stratified TCGA-derived cases as a low- or high-risk group, whereas the significance of prognosis was verified based on CGGA-derived cases. Kaplan-Meier analysis revealed that the high-risk group was associated with the dismal prognosis. Furthermore, the S100 family-based signature was proved to be closely related to immune microenvironment. In vitro analysis showed S100B gene in the signature promoted glioblastoma (GBM) cell proliferation and migration. Conclusions We constructed and verified a novel S100 family-based signature associated with tumor immune microenvironment (TIME), which may shed novel light on the glioma diagnosis and treatment.
Collapse
|
6
|
Zhang DY, Monteiro MJ, Liu JP, Gu WY. Mechanisms of cancer stem cell senescence: Current understanding and future perspectives. Clin Exp Pharmacol Physiol 2021; 48:1185-1202. [PMID: 34046925 DOI: 10.1111/1440-1681.13528] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 05/24/2021] [Indexed: 12/13/2022]
Abstract
Cancer stem cells (CSCs) are a small population of heterogeneous tumor cells with the capacity of self-renewal and aberrant differentiation for immortality and divergent lineages of cancer cells. In contrast to bulky tumor cells, CSCs remain less differentiated and resistant to therapy even when targeted with tissue-specific antigenic markers. This makes CSCs responsible for not only tumor initiation, development, but also tumor recurrence. Emerging evidence suggests that CSCs can undergo cell senescence, a non-proliferative state of cells in response to stress. While cell senescence attenuates tumor cell proliferation, it is commonly regarded as a tumor suppressive mechanism. However, mounting research indicates that CSC senescence also provides these cells with the capacity to evade cytotoxic effects from cancer therapy, exacerbating cancer relapse and metastasis. Recent studies demonstrate that senescence drives reprogramming of cancer cell toward stemness and promotes CSC generation. In this review, we highlight the origin, heterogeneity and senescence regulatory mechanisms of CSCs, the complex relationship between CSC senescence and tumor therapy, and the recent beneficial effects of senotherapy on eliminating senescent tumor cells.
Collapse
Affiliation(s)
- Da-Yong Zhang
- Department of Clinical Medicine, Zhejiang University City College, Hangzhou, China
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, Australia
| | - Michael J Monteiro
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, Australia
| | - Jun-Ping Liu
- Institute of Ageing Research, Hangzhou Normal University, Hangzhou, China
- Department of Immunology, Monash University Faculty of Medicine, Prahran, Vic, Australia
- Hudson Institute of Medical Research, and Department of Molecular and Translational Science, Monash University Faculty of Medicine, Clayton, Vic, Australia
| | - Wen-Yi Gu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
7
|
Tian Y, Cao R, Che B, Sun D, Tang Y, Jiang L, Bai Q, Liu Y, Morozova-Roche LA, Zhang C. Proinflammatory S100A9 Regulates Differentiation and Aggregation of Neural Stem Cells. ACS Chem Neurosci 2020; 11:3549-3556. [PMID: 33079539 DOI: 10.1021/acschemneuro.0c00365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inflammation is the primary pathological feature of neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease. Proinflammatory molecules (e.g., S100A9) play important roles during the progression of the diseases by regulating behavior and fate of multiple cell types in the nervous system. Our earlier studies reveal that S100A9 is toxic to neurons, and its interaction with Aβ peptides leads to the formation of large nontoxic amyloidogenic aggregates, suggesting a protective role of coaggregation with Aβ amyloids. We herein demonstrate that S100A9 interacts with neural stem cells (NSCs) and causes NSC differentiation. In the brain of transgenic AD mouse models, we found large quantities of proinflammatory S100A9, which colocalizes with the differentiated NSCs. NSC sphere formation, which is a representative character of NSC stemness, is also substantially inhibited by S100A9. These results suggest that S100A9 is a representative marker for the inflammatory conditions in AD, and it promotes NSC differentiation. Intriguingly, in contrast to the death of both stem and differentiated NSCs caused by high S100A9 doses, S100A9 at a moderate concentration is toxic only to the early differentiated NSCs but not the stem cells. We therefore postulate that, at the early stage of AD, the expression of S100A9 leads to NSC differentiation, which remedies the neuron damage. The application of drugs, which help maintain NSC stemness (e.g., the platelet-derived growth factor, PDGF), may help overcome the acute inflammatory conditions and improve the efficacy of NSC transplantation therapy.
Collapse
Affiliation(s)
- Yin Tian
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University & Institute of Photonics and Photon-Technology, Northwest University, 1 Xue Fu Avenue, Xi’an, Shaanxi 710127, China
| | - Rui Cao
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University & Institute of Photonics and Photon-Technology, Northwest University, 1 Xue Fu Avenue, Xi’an, Shaanxi 710127, China
| | - Bingchen Che
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University & Institute of Photonics and Photon-Technology, Northwest University, 1 Xue Fu Avenue, Xi’an, Shaanxi 710127, China
| | - Dan Sun
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University & Institute of Photonics and Photon-Technology, Northwest University, 1 Xue Fu Avenue, Xi’an, Shaanxi 710127, China
| | - Yong Tang
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University & Institute of Photonics and Photon-Technology, Northwest University, 1 Xue Fu Avenue, Xi’an, Shaanxi 710127, China
| | - Lin Jiang
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University & Institute of Photonics and Photon-Technology, Northwest University, 1 Xue Fu Avenue, Xi’an, Shaanxi 710127, China
| | - Qiao Bai
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University & Institute of Photonics and Photon-Technology, Northwest University, 1 Xue Fu Avenue, Xi’an, Shaanxi 710127, China
| | - Yonggang Liu
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University & Institute of Photonics and Photon-Technology, Northwest University, 1 Xue Fu Avenue, Xi’an, Shaanxi 710127, China
| | | | - Ce Zhang
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University & Institute of Photonics and Photon-Technology, Northwest University, 1 Xue Fu Avenue, Xi’an, Shaanxi 710127, China
- Department of Pharmacy, Chongqing Three Gorges Medical College, Chongqing 404120, China
| |
Collapse
|
8
|
Tian Z, Tang J, Liao X, Yang Q, Wu Y, Wu G. An immune-related prognostic signature for predicting breast cancer recurrence. Cancer Med 2020; 9:7672-7685. [PMID: 32841536 PMCID: PMC7571818 DOI: 10.1002/cam4.3408] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/25/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BC) is the most common cancer among women worldwide and is the second leading cause of cancer‐related deaths in women. Increasing evidence has validated the vital role of the immune system in BC development and recurrence. In this study, we identified an immune‐related prognostic signature of BRCA that could help delineate risk scores of poor outcome for each patient. This prognostic signature comprised information on five danger genes—TSLP, BIRC5, S100B, MDK, and S100P—and three protect genes RARRES3, BLNK, and ACO1. Kaplan‐Meier survival curve showed that patients classified as low‐risk according to optimum cut‐off risk score had better prognosis than those identified within the high‐risk group. ROC analysis indicated that the identified prognostic signature had excellent diagnostic efficiency for predicting 3‐ and 5‐years relapse‐free survival (RFS). Multivariate Cox regression analysis proved that the prognostic signature is independent of other clinical parameters. Stratification analysis demonstrated that the prognostic signature can be used to predict the RFS of BC patients within the same clinical subgroup. We also developed a nomogram to predict the RFS of patients. The calibration plots exhibited outstanding performance. The validation sets (GSE21653, GSE20711, and GSE88770) were used to external validation. More convincingly, the real time RT‐PCR results of clinical samples demonstrated that danger genes were significantly upregulated in BC samples, whereas protect genes were downregulated. In conclusion, we developed and validated an immune‐related prognostic signature, which exhibited excellent diagnostic efficiency in predicting the recurrence of BC, and will help to make personalized treatment decisions for patients at different risk score.
Collapse
Affiliation(s)
- Zelin Tian
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jianing Tang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xing Liao
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qian Yang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yumin Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Gaosong Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Allgöwer C, Kretz AL, von Karstedt S, Wittau M, Henne-Bruns D, Lemke J. Friend or Foe: S100 Proteins in Cancer. Cancers (Basel) 2020; 12:cancers12082037. [PMID: 32722137 PMCID: PMC7465620 DOI: 10.3390/cancers12082037] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/24/2022] Open
Abstract
S100 proteins are widely expressed small molecular EF-hand calcium-binding proteins of vertebrates, which are involved in numerous cellular processes, such as Ca2+ homeostasis, proliferation, apoptosis, differentiation, and inflammation. Although the complex network of S100 signalling is by far not fully deciphered, several S100 family members could be linked to a variety of diseases, such as inflammatory disorders, neurological diseases, and also cancer. The research of the past decades revealed that S100 proteins play a crucial role in the development and progression of many cancer types, such as breast cancer, lung cancer, and melanoma. Hence, S100 family members have also been shown to be promising diagnostic markers and possible novel targets for therapy. However, the current knowledge of S100 proteins is limited and more attention to this unique group of proteins is needed. Therefore, this review article summarises S100 proteins and their relation in different cancer types, while also providing an overview of novel therapeutic strategies for targeting S100 proteins for cancer treatment.
Collapse
Affiliation(s)
- Chantal Allgöwer
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (C.A.); (A.-L.K.); (M.W.); (D.H.-B.)
| | - Anna-Laura Kretz
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (C.A.); (A.-L.K.); (M.W.); (D.H.-B.)
| | - Silvia von Karstedt
- Department of Translational Genomics, Center of Integrated Oncology Cologne-Bonn, Medical Faculty, University Hospital Cologne, Weyertal 115b, 50931 Cologne, Germany;
- CECAD Cluster of Excellence, University of Cologne, Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany
- Center of Molecular Medicine Cologne, Medical Faculty, University Hospital of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - Mathias Wittau
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (C.A.); (A.-L.K.); (M.W.); (D.H.-B.)
| | - Doris Henne-Bruns
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (C.A.); (A.-L.K.); (M.W.); (D.H.-B.)
| | - Johannes Lemke
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (C.A.); (A.-L.K.); (M.W.); (D.H.-B.)
- Correspondence: ; Tel.: +49-731-500-53691
| |
Collapse
|
10
|
Wu KJ, Wang W, Wang HMD, Leung CH, Ma DL. Interfering with S100B-effector protein interactions for cancer therapy. Drug Discov Today 2020; 25:1754-1761. [PMID: 32679172 DOI: 10.1016/j.drudis.2020.07.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/17/2020] [Accepted: 07/02/2020] [Indexed: 12/17/2022]
Abstract
S100 calcium-binding protein B (S100B) is overexpressed in various malignant tumors, where it regulates cancer cell proliferation and metabolism by physical interactions with other molecules. Interfering with S100B-effector protein interactions is a potential strategy to treat malignant tumors. Although some S100B inhibitors have been discovered by virtual screening (VS), most target the S100B-p53 interaction. Hence, there is scope for the discovery of other S100B-effector protein interaction modulators for malignant tumors. In this review, we provide an overview of S100B-effector protein interaction inhibitor discovery using VS and discuss promising S100B-effector protein interaction targets that permit in silico analysis for drug discovery.
Collapse
Affiliation(s)
- Ke-Jia Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa 999078, Macao SAR, China
| | - Wanhe Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong, China
| | - Hui-Min David Wang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung 402, Taiwan
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa 999078, Macao SAR, China.
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong, China.
| |
Collapse
|
11
|
El-Far AH, Sroga G, Al Jaouni SK, Mousa SA. Role and Mechanisms of RAGE-Ligand Complexes and RAGE-Inhibitors in Cancer Progression. Int J Mol Sci 2020; 21:ijms21103613. [PMID: 32443845 PMCID: PMC7279268 DOI: 10.3390/ijms21103613] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/02/2020] [Accepted: 05/08/2020] [Indexed: 12/26/2022] Open
Abstract
Interactions of the receptor for advanced glycation end product (RAGE) and its ligands in the context of their role in diabetes mellitus, inflammation, and carcinogenesis have been extensively investigated. This review focuses on the role of RAGE-ligands and anti-RAGE drugs capable of controlling cancer progression. Different studies have demonstrated interaction of RAGE with a diverse range of acidic (negatively charged) ligands such as advanced glycation end products (AGEs), high-mobility group box1 (HMGB1), and S100s, and their importance to cancer progression. Some RAGE-ligands displayed effects on anti- and pro-apoptotic proteins through upregulation of the phosphatidylinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR), mitogen-activated protein kinases (MAPKs), matrix metalloproteinases (MMPs), vascular endothelial growth factor (VEGF), and nuclear factor kappa B (NF-κB) pathways, while downregulating p53 in cancer progression. In addition, RAGE may undergo ligand-driven multimodal dimerization or oligomerization mediated through self-association of some of its subunits. We conclude our review by proposing possible future lines of study that could result in control of cancer progression through RAGE inhibition.
Collapse
Affiliation(s)
- Ali H. El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Damanhour 22511, Egypt;
| | - Grazyna Sroga
- Rensselaer Polytechnic Institute, NY (RPI), Troy, NY 12180, USA;
| | - Soad K. Al Jaouni
- Department of Hematology/Pediatric Oncology, King Abdulaziz University, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Shaker A. Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA
- Correspondence:
| |
Collapse
|
12
|
Hua X, Zhang H, Jia J, Chen S, Sun Y, Zhu X. Roles of S100 family members in drug resistance in tumors: Status and prospects. Biomed Pharmacother 2020; 127:110156. [PMID: 32335300 DOI: 10.1016/j.biopha.2020.110156] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023] Open
Abstract
Chemotherapy and targeted therapy can significantly improve survival rates in cancer, but multiple drug resistance (MDR) limits the efficacy of these approaches. Understanding the molecular mechanisms underlying MDR is crucial for improving drug efficacy and clinical outcomes of patients with cancer. S100 proteins belong to a family of calcium-binding proteins and have various functions in tumor development. Increasing evidence demonstrates that the dysregulation of various S100 proteins contributes to the development of drug resistance in tumors, providing a basis for the development of predictive and prognostic biomarkers in cancer. Therefore, a combination of biological inhibitors or sensitizers of dysregulated S100 proteins could enhance therapeutic responses. In this review, we provide a detailed overview of the mechanisms by which S100 family members influence resistance of tumors to cancer treatment, with a focus on the development of effective strategies for overcoming MDR.
Collapse
Affiliation(s)
- Xin Hua
- Southeast University Medical College, Nanjing, 210009, China.
| | - Hongming Zhang
- Department of Respiratory Medicine, Yancheng Third People's Hospital, Southeast University Medical College, Yancheng, 224000, China.
| | - Jinfang Jia
- Southeast University Medical College, Nanjing, 210009, China.
| | - Shanshan Chen
- Southeast University Medical College, Nanjing, 210009, China.
| | - Yue Sun
- Southeast University Medical College, Nanjing, 210009, China.
| | - Xiaoli Zhu
- Southeast University Medical College, Nanjing, 210009, China; Department of Respiratory Medicine, Zhongda Hospital of Southeast University Medical College, Nanjing, 210009, China.
| |
Collapse
|
13
|
Riuzzi F, Chiappalupi S, Arcuri C, Giambanco I, Sorci G, Donato R. S100 proteins in obesity: liaisons dangereuses. Cell Mol Life Sci 2020; 77:129-147. [PMID: 31363816 PMCID: PMC11104817 DOI: 10.1007/s00018-019-03257-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/19/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023]
Abstract
Obesity is an endemic pathophysiological condition and a comorbidity associated with hypercholesterolemia, hypertension, cardiovascular disease, type 2 diabetes mellitus, and cancer. The adipose tissue of obese subjects shows hypertrophic adipocytes, adipocyte hyperplasia, and chronic low-grade inflammation. S100 proteins are Ca2+-binding proteins exclusively expressed in vertebrates in a cell-specific manner. They have been implicated in the regulation of a variety of functions acting as intracellular Ca2+ sensors transducing the Ca2+ signal and extracellular factors affecting cellular activity via ligation of a battery of membrane receptors. Certain S100 proteins, namely S100A4, the S100A8/S100A9 heterodimer and S100B, have been implicated in the pathophysiology of obesity-promoting macrophage-based inflammation via toll-like receptor 4 and/or receptor for advanced glycation end-products ligation. Also, serum levels of S100A4, S100A8/S100A9, S100A12, and S100B correlate with insulin resistance/type 2 diabetes, metabolic risk score, and fat cell size. Yet, secreted S100B appears to exert neurotrophic effects on sympathetic fibers in brown adipose tissue contributing to the larger sympathetic innervation of this latter relative to white adipose tissue. In the present review we first briefly introduce S100 proteins and then critically examine their role(s) in adipose tissue and obesity.
Collapse
Affiliation(s)
- Francesca Riuzzi
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
| | - Sara Chiappalupi
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
| | - Cataldo Arcuri
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Ileana Giambanco
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Guglielmo Sorci
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
- Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, 06132, Perugia, Italy
| | - Rosario Donato
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy.
| |
Collapse
|
14
|
Ma N, Zhu L, Yang L, Cui Y, Zhan Y. Prognostic values of S100 family mRNA expression in ovarian cancer. Cancer Biomark 2019; 25:67-78. [PMID: 31033462 DOI: 10.3233/cbm-182276] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
S100 family is made up of at least 20 calcium-binding proteins which are involved in many cellular processes. The prognostic values of individual S100 member in ovarian cancer patients are still unknown. In this study, we performed a detailed prognostic values of S100 in ovarian cancer. The mRNA expression levels of S100 family in various cancers were analyzed via the Oncomine, and the protein-protein interaction network of S100 family was analyzed via String. The prognostic values of individual S100 member were evaluated via Kaplan-Meier Plotter. The S100 family genes expression and mutation were analyzed via cBioProtal. We observed that the mRNA expression of most S100 family were overexpressed in ovarian cancer compared with normal tissues. In survival analysis in Kaplan-Meier Plotter, 10 members of S100 family showed significant correlation with overall survival in ovarian cancer patients. The trends of high expression of individual S100 members were nearly the same in different subtype and pathological grades. However, the S100 family genes expression and mutation showed no significant prognostic values in overall survival and disease free survival in ovarian cancer patients. Although, the results need more verification both in clinical trials and fundamental experiments, our study provide new insights for the prognostic function of S100 family in ovarian cancer and might promote development of S100 targeted inhibitors for the new treatment of ovarian cancer.
Collapse
Affiliation(s)
- Nan Ma
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lizhe Zhu
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Liu Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuxin Cui
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yingzhuan Zhan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
15
|
Wu Y, Liu X, Guo LY, Zhang L, Zheng F, Li S, Li XY, Yuan Y, Liu Y, Yan YW, Chen SY, Wang JN, Zhang JX, Tang JM. S100B is required for maintaining an intermediate state with double-positive Sca-1+ progenitor and vascular smooth muscle cells during neointimal formation. Stem Cell Res Ther 2019; 10:294. [PMID: 31547879 PMCID: PMC6757428 DOI: 10.1186/s13287-019-1400-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022] Open
Abstract
Introduction Accumulation of vascular smooth muscle cells (VSMCs) within the neointimal region is a hallmark of atherosclerosis and vessel injury. Evidence has shown that Sca-1-positive (Sca-1+) progenitor cells residing in the vascular adventitia play a crucial role in VSMC assemblages and intimal lesions. However, the underlying mechanisms, especially in the circumstances of vascular injury, remain unknown. Methods and results The neointimal formation model in rats was established by carotid artery balloon injury using a 2F-Forgaty catheter. Most Sca-1+ cells first appeared at the adventitia of the vascular wall. S100B expressions were highest within the adventitia on the first day after vessel injury. Along with the sequentially increasing trend of S100B expression in the intima, media, and adventitia, respectively, the numbers of Sca-1+ cells were prominently increased at the media or neointima during the time course of neointimal formation. Furthermore, the Sca-1+ cells were markedly increased in the tunica media on the third day of vessel injury, SDF-1α expressions were obviously increased, and SDF-1α levels and Sca-1+ cells were almost synchronously increased within the neointima on the seventh day of vessel injury. These effects could effectually be reversed by knockdown of S100B by shRNA, RAGE inhibitor (SPF-ZM1), or CXCR4 blocker (AMD3100), indicating that migration of Sca-1+ cells from the adventitia into the neointima was associated with S100B/RAGE and SDF-1α/CXCR4. More importantly, the intermediate state of double-positive Sca-1+ and α-SMA cells was first found in the neointima of injured arteries, which could be substantially abrogated by using shRNA for S100B or blockade of CXCR4. S100B dose-dependently regulated SDF-1α expressions in VSMCs by activating PI3K/AKT and NF-κB, which were markedly abolished by PI3K/AKT inhibitor wortmannin and enhanced by p65 blocker PDTC. Furthermore, S100B was involved in human umbilical cord-derived Sca-1+ progenitor cells’ differentiation into VSMCs, especially in maintaining the intermediate state of double-positive Sca-1+ and α-SMA. Conclusions S100B triggered neointimal formation in rat injured arteries by maintaining the intermediate state of double-positive Sca-1+ progenitor and VSMCs, which were associated with direct activation of RAGE by S100B and indirect induction of SDF-1α by activating PI3K/AKT and NF-κB. Electronic supplementary material The online version of this article (10.1186/s13287-019-1400-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yan Wu
- Department of Physiology, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Xin Liu
- Laboratory Animal Center, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Ling-Yun Guo
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Lei Zhang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Fei Zheng
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Shan Li
- Department of Biochemistry, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Xing-Yuan Li
- Department of Physiology, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Ye Yuan
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Yu Liu
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Yu-Wen Yan
- Department of Physiology, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Shi-You Chen
- Department of Physiology & Pharmacology, The University of Georgia, Athens, GA, 30602, USA
| | - Jia-Ning Wang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Jin-Xuan Zhang
- Department of Physiology, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China. .,Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| | - Jun-Ming Tang
- Department of Physiology, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China. .,Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China. .,Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| |
Collapse
|
16
|
Bai Y, Li LD, Li J, Lu X. Prognostic values of S100 family members in ovarian cancer patients. BMC Cancer 2018; 18:1256. [PMID: 30558666 PMCID: PMC6296138 DOI: 10.1186/s12885-018-5170-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 12/02/2018] [Indexed: 01/06/2023] Open
Abstract
Objective Exhibiting high consistence in sequence and structure, S100 family members are interchangeable in function and they show a wide spectrum of biological processes, including proliferation, apoptosis, migration, inflammation and differentiation and the like. While the prognostic value of each individual S100 in ovarian cancer is still elusive. In current study, we investigated the prognostic value of S100 family members in the ovarian cancer. Methods We used the Kaplan Meier plotter (KM plotter) database, in which updated gene expression data and survival information are from 1657 ovarian cancer patients, to assess the relevance of individual S100 family mRNA expression to overall survival in various ovarian cancer subtypes and different clinicopathological features. Results It was found that high expression of S100A2 (HR = 1.18, 95%CI: 1.04–1.34, P = 0.012), S100A7A (HR = 1.3, 95%CI: 1.04–1.63, P = 0.02),S100A10 (HR = 1.2, 95%CI: 1.05–1.38, P = 0.0087),and S100A16 (HR = 1.23, 95%CI: 1–1.51, P = 0.052) were significantly correlated with worse OS in all ovarian cancer patients, while the expression of S100A1 (HR = 0.87, 95%CI: 0.77–0.99, P = 0.039), S100A3 (HR = 0.83, 95%CI: 0.71–0.96, P = 0.0011), S100A5 (HR = 0.84, 95%CI: 0.73–0.97, P = 0.017), S100A6 (HR = 0.84, 95%CI: 0.72–0.98, P = 0.024), S100A13 (HR = 0.85, 95%CI:0.75–0.97, P = 0.014) and S100G (HR = 0.86, 95%CI: 0.74–0.99, P = 0.041) were associated with better prognosis. Furthermore, we assessed the prognostic value of S100 expression in different subtypes and the clinicopathological features, including pathological grades, clinical stages and TP53 mutation status, of ovarian cancer patients. Conclusion Comprehensive understanding of the S100 family members may have guiding significance for the diagnosis and outcome of ovarian cancer patients. Electronic supplementary material The online version of this article (10.1186/s12885-018-5170-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yang Bai
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China.,Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Liang-Dong Li
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, 200030, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200030, China
| | - Jun Li
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China.,Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Xin Lu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China. .,Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China. .,Present Address: Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, No.419, Fangxie Road, Shanghai, 200011, China.
| |
Collapse
|
17
|
Al-Alem LF, Pandya UM, Baker AT, Bellio C, Zarrella BD, Clark J, DiGloria CM, Rueda BR. Ovarian cancer stem cells: What progress have we made? Int J Biochem Cell Biol 2018; 107:92-103. [PMID: 30572025 DOI: 10.1016/j.biocel.2018.12.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/14/2018] [Accepted: 12/16/2018] [Indexed: 12/18/2022]
Abstract
Ovarian cancer (OvCa) is the most lethal gynecological malignancy in the United States primarily due to lack of a reliable early diagnostic, high incidence of chemo-resistant recurrent disease as well as profuse tumor heterogeneity. Cancer stem cells (CSCs) continue to gain attention, as they are known to resist chemotherapy, self-renew and re-populate the bulk tumor with undifferentiated and differentiated cells. Moreover, CSCs appear to readily adapt to environmental, immunologic and pharmacologic cues. The plasticity and ability to inactivate or activate signaling pathways promoting their longevity has been, and continues to be, the challenge faced in developing successful CSC targeted therapies. Identifying and understanding unique ovarian CSC markers and the pathways they utilize could reveal new therapeutic opportunities that may offer alternative adjuvant treatment options. Herein, we will discuss the current state of ovarian CSC characterization, their contribution to disease resistance, recurrence and shed light on clinical trials that may target the CSC population.
Collapse
Affiliation(s)
- Linah F Al-Alem
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Unnati M Pandya
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Andrew T Baker
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Chiara Bellio
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Bianca D Zarrella
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Celeste M DiGloria
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
| | - Bo R Rueda
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Ultrasound microbubbles mediated miR-let-7b delivery into CD133 + ovarian cancer stem cells. Biosci Rep 2018; 38:BSR20180922. [PMID: 30126854 PMCID: PMC6165842 DOI: 10.1042/bsr20180922] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/19/2018] [Accepted: 08/07/2018] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer stem cells (OCSCs) are considered the reason for ovarian cancer’s emergence and recurrence. Ultrasound-targetted microbubble destruction (UTMD), a non-vial, safe, and promising delivery method for miRNA, is reported to transfect cancer stem cells (CSCs). In the present study, we investigated to transfect miR-let-7b into OCSCs using UTMD. The CD133+ OCSCs, accounted for only 0.1% of ovarian cancer cell line A2780, were separated by flow cytometry, and the CSC characteristics of CD133+ OCSCs have been proved by spheroid formation and self-renewal assay. The miR-let-7b transfection efficiency using UTMD was significantly higher than other groups except lipofectamine group through flow cytometry. The cell viability of all groups decreased after transfection, and the late apoptosis rate of CD133+ OCSCs after miR-let7b transfection induced by UTMD was 2.62%, while that of non-treated cells was 0.02% (P<0.05). Furthermore, the Western blot results demonstrated that the stem cells surface marker of CD133 expression has decreased. Therefore, our results indicated that UTMD-mediated miRNA delivery could be a promising platform for CSC therapy.
Collapse
|
19
|
Dai CX, Hu CC, Shang YS, Xie J. Role of Ginkgo biloba extract as an adjunctive treatment of elderly patients with depression and on the expression of serum S100B. Medicine (Baltimore) 2018; 97:e12421. [PMID: 30278520 PMCID: PMC6181482 DOI: 10.1097/md.0000000000012421] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVE To explore the effect of ginkgo biloba extract (EGb) as an adjunctive treatment of elderly patients with depression and the effect on the expression of serum S100B. METHODS 136 elderly patients with depression were divided into EGb + citalopram (Cit) group and Cit group equally. Efficacy was evaluated by Hamilton Depression Rating Scale (HAMD). Wisconsin Card Classification Test (WCST) was used to evaluate cognitive function. Serum S100B expression was measured with ELISA. The relationship of S100B with HAMD, Hamilton Anxiety Scale (HAMA) score, and WCST results was evaluated subsequently. RESULTS The time of onset of efficacy was significantly shorter in EGb + Cit group. There were significant differences in HAMD and HAMA scores after treatment than before treatment between groups (all P < .05). After treatment, total number of WCST test, the number of continuous errors and non-persistent errors in both groups were less than those before treatment. The correct number and classifications number were increased than before treatment. In EGb + Cit group, correct numbers and classifications were increased, and the number of persistent errors was decreased. After treatment, S100B level was decreased, and S100B levels change in EGb + Cit group was greater than in Cit group. Serum S100B level was positively correlated with HAMD and HAMA scores before treatment and positively correlated with persistent errors number in WCST. CONCLUSION EGb, as an adjunctive treatment, can effectively improve depressive symptoms and reduce expression of serum S100B, which is a marker of brain injury, suggesting that EGb restores neurologic function during the treatment of depression in elderly patients and S100B participates in the therapeutic mechanism. EGb combined with depressive drugs plays synergistic role, and the time of onset of efficacy is faster than single antidepressants.
Collapse
|
20
|
Ermakov A, Daks A, Fedorova O, Shuvalov O, Barlev NA. Ca 2+ -depended signaling pathways regulate self-renewal and pluripotency of stem cells. Cell Biol Int 2018; 42:1086-1096. [PMID: 29851182 DOI: 10.1002/cbin.10998] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 05/25/2018] [Indexed: 12/15/2022]
Abstract
Ca2+ -mediated signaling is widely spread in nature and plays critical role in the individual development of various organisms ranging from microorganisms to mammals. In vertebrates, Ca2+ is involved in important developmental events: fertilization, body plan establishment, and organogenesis. The two later events are defined by embryonic stem cells (ESCs). ESCs are capable of self-renewal and are pluripotent by nature, that is, can give rise to all types of cells that make up the body. Given the paramount importance of Ca2+ signalization in the development, it is therefore not surprising this process also plays role in the biology of stem cells. In this review, we scrutinize the published experimental data on the role of Ca2+ ions in embryonic stem cells self-renewal and pluripotency. In line with this, we also discuss possible mechanisms of p53 inhibition as a major hindrance to self-renewal of ESCs. Finally, we argue about the role of G-protein-coupled receptors (GPCRs), the largest family of heteromeric transmembrane receptors, and GPCR-mediated signalization in stem cells, and propose the role for the GPCR-G-protein-PLC-Ca2+ -downstream signaling pathway in the regulation of pluripotency of both mouse and human ESCs.
Collapse
Affiliation(s)
| | - Alexandra Daks
- Institute of Cytology RAS, Saint-Petersburg 194064, Russia
| | - Olga Fedorova
- Institute of Cytology RAS, Saint-Petersburg 194064, Russia
| | - Oleg Shuvalov
- Institute of Cytology RAS, Saint-Petersburg 194064, Russia
| | | |
Collapse
|
21
|
Raffat MA, Hadi NI, Hosein M, Mirza S, Ikram S, Akram Z. S100 proteins in oral squamous cell carcinoma. Clin Chim Acta 2018; 480:143-149. [DOI: 10.1016/j.cca.2018.02.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/11/2018] [Accepted: 02/13/2018] [Indexed: 01/11/2023]
|
22
|
Yen MC, Huang YC, Kan JY, Kuo PL, Hou MF, Hsu YL. S100B expression in breast cancer as a predictive marker for cancer metastasis. Int J Oncol 2017; 52:433-440. [PMID: 29345293 DOI: 10.3892/ijo.2017.4226] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/05/2017] [Indexed: 11/06/2022] Open
Abstract
In the tumor microenvironment, soluble molecules play important role in the establishment of a pre-metastatic niche. The S100 calcium-binding protein family are inflammatory molecules that contribute to the development of a pro-inflammatory tumor microenvironment. S100B belongs to the S100 family and serum S100B (also known as S100beta) serves as a marker for metastasis in lung cancer, ovarian cancer and melanoma. However, the association between S100B and the metastasis of breast cancer is not yet well understood. In the present study, a relatively low S100B expression was observed in the tumor samples compared to normal breast tissue among online microarray datasets. When the estrogen receptor (ER)-negative breast cancer cell lines, MDA-MB-231 and Hs578T, were treated with recombinant human S100B, cell migration was significantly inhibited and epithelial cadherin expression was increased. Our results revealed that a high S100B expression predicted a good overall survival in patients with ER-negative breast cancer, and good distant metastases-free survival in all patients with breast cancer via the analysis of the KM plotter and SurvExpress databases. Although previous studies have indicated that the interaction of S100B with wild-type p53 inhibits p53 function, a high S100B expression is associated with a good prognosis in patients with p53 mutant and p53 wild-type breast cancers. On the whole, our findings demonstrate that S100B treatment suppresses the migratory capacity of ER-negative breast cancer and that S100B expression may serve a predictive marker for metastasis in breast cancer.
Collapse
Affiliation(s)
- Meng-Chi Yen
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Yung-Chi Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Jung-Yu Kan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Ming-Feng Hou
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Ya-Ling Hsu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| |
Collapse
|
23
|
Nolta JA. Research Leads to Approved Therapies in the New Era of Living Medicine. Stem Cells 2017; 36:1-3. [PMID: 29210150 DOI: 10.1002/stem.2748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 11/28/2017] [Indexed: 11/08/2022]
|
24
|
Tan B, Shen L, Yang K, Huang D, Li X, Li Y, Zhao L, Chen J, Yi Q, Xu H, Tian J, Zhu J. C6 glioma-conditioned medium induces malignant transformation of mesenchymal stem cells: Possible role of S100B/RAGE pathway. Biochem Biophys Res Commun 2017; 495:78-85. [PMID: 29050939 DOI: 10.1016/j.bbrc.2017.10.071] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/15/2017] [Indexed: 01/03/2023]
Abstract
Mesenchymal stem cells (MSCs) have been widely studied as an attractive therapeutic agent for the treatment of tumors. However, the adverse effects of the tumor paracrine factors who affect MSCs are still unclear. In this study, we report for the first time that C6 glioma-conditioned medium (GCM) induces malignant transformation of MSCs. In contrast to MSCs, the transformed mesenchymal stem cells (TMCs) exhibited tumor cell characterizations in vitro and highly tumorigenic in vivo. Furthermore, GCM and recombinant S100B increased receptor for advanced glycation end products (RAGE) and its downstream Akt1, STAT3 genes expression as well as phosphorylation and transcriptional activation. Finally, blockage of S100B-RAGE interaction by RAGE inhibitor FPS-ZM1 attenuated GCM and S100B-induced Akt1, STAT3 activation, abolished its cell proliferation, migration and invasion actions. Together, these results suggest that the RAGE pathway may play a possible role in malignant transformation procedure of MSCs, and that this process may be mediated through S100B.
Collapse
Affiliation(s)
- Bin Tan
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China
| | - Lianju Shen
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, China
| | - Ke Yang
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, 400014, China
| | - Daochao Huang
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China
| | - Xin Li
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China
| | - Yasha Li
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China
| | - Li Zhao
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China
| | - Jie Chen
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China
| | - Qing Yi
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China
| | - Hao Xu
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China
| | - Jie Tian
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China
| | - Jing Zhu
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China.
| |
Collapse
|
25
|
Zhang S, Wang Z, Liu W, Lei R, Shan J, Li L, Wang X. Distinct prognostic values of S100 mRNA expression in breast cancer. Sci Rep 2017; 7:39786. [PMID: 28051137 PMCID: PMC5209742 DOI: 10.1038/srep39786] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/28/2016] [Indexed: 12/22/2022] Open
Abstract
S100 family genes encode low molecular weight, acidic-Ca2+ binding proteins implicating in a wide spectrum of biological processes. S100 family contains at least 20 members, most of which are frequently dysregulated in human malignancies including breast cancer. However, the prognostic roles of each individual S100, especially the mRNA level, in breast cancer patients remain elusive. In the current study, we used "The Kaplan-Meier plotter" (KM plotter) database to investigate the prognostic values of S100 mRNA expression in breast cancer. Our results indicated that high mRNA expression of S100A8, S100A9, S100A11 and S100P were found to be significantly correlated to worse outcome, while S100A1 and S100A6 were associated with better prognosis in all breast cancer patients. We further assessed the prognostic value of S100 in different intrinsic subtypes and clinicopathological features of breast cancer. The associated results will elucidate the role of S100 in breast cancer and may further lead the research to explore the S100-targeting reagents for treating breast cancer patients.
Collapse
Affiliation(s)
- Shizhen Zhang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou, Zhejiang 310009, China.,Cancer Institute (Key Laboratory of Cancer Prevention &Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences), Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Zhen Wang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou, Zhejiang 310009, China.,Cancer Institute (Key Laboratory of Cancer Prevention &Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences), Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Weiwei Liu
- Department of Laboratory Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Rui Lei
- Department of Plastic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, No. 79, Qingchun Road, Hangzhou, Zhejiang 310009, China
| | - Jinlan Shan
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou, Zhejiang 310009, China.,Cancer Institute (Key Laboratory of Cancer Prevention &Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences), Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Ling Li
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Xiaochen Wang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou, Zhejiang 310009, China.,Cancer Institute (Key Laboratory of Cancer Prevention &Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences), Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88, Jiefang Road, Hangzhou, Zhejiang 310009, China
| |
Collapse
|