1
|
Zhang Y, Zhao J, Guo H, Lu X, Tan D. Production and Bioseparation Applications of Polyhydroxyalkanoate Nano-Granules Functionalized with Streptavidin. Microorganisms 2025; 13:312. [PMID: 40005680 PMCID: PMC11858450 DOI: 10.3390/microorganisms13020312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Rapidly growing industrial biotechnology and bio-manufacturing require simple and cost-effective bioseparation tools. A novel strategy of bioseparation based on the streptavidin-decorated polyhydroxyalkanoate (PHA) nano-granules was developed in this study. By fusing to the N-terminus of PHA-associated phasin protein, the streptavidin was one-step immobilized on the surface of PHA nano-granules simultaneously with the accumulation of PHA in recombinant Escherichia coli. About 1.95 g/L of PHA nano-granules (54.51 wt% of cell dry weight) were produced after 48 h bacterial cultivation. The following qualitative and quantitative characterizations demonstrated that the streptavidin accounted for approximately 6.78% of the total weight of the purified PHA nano-granules and confirmed a considerable biotin affinity of 0.1 ng biotin/μg surface protein. As a proof of concept, the nano-granules were further functionalized with biotinylated oligo(dT) for mRNA isolation and about 1.26 μg of mRNA (occupied 2.59%) was purified from 48.45 μg of total RNA, achieving good integrity and high purity with few DNA and rRNA contaminations. Moreover, the nano-granules retained more than 80% of their initial mRNA recovery efficiency after ten cycles of repeated use. The PHA-SAP nano-granules were also functionalized with biotinylated magnetic beads, allowing magnetic recovery of the PHA nano-granules from cell lysates that still needs optimization. Our study provides a novel and expandable platform of PHA nano-granules that can be further functionalized with various biological groups for bioseparation applications. The functional PHA nano-granules have a great potential to serve as bioseparation resin for large-scale purification processes after suitable optimizations for "bench-to-factory" translation, contributing to scalable and sustainable bioprocessing.
Collapse
Affiliation(s)
- Yuyan Zhang
- School of Life Science & Technology, Xinjiang University, Urumchi 830049, China;
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biological Science and Bioengineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (J.Z.); (H.G.)
| | - Jiping Zhao
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biological Science and Bioengineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (J.Z.); (H.G.)
| | - Hui Guo
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biological Science and Bioengineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (J.Z.); (H.G.)
| | - Xiaoyun Lu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biological Science and Bioengineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (J.Z.); (H.G.)
| | - Dan Tan
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biological Science and Bioengineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (J.Z.); (H.G.)
| |
Collapse
|
2
|
Pietrobono D, Russo L, Bertilacchi MS, Marchetti L, Martini C, Giacomelli C, Trincavelli ML. Extracellular adenosine oppositely regulates the purinome machinery in glioblastoma and mesenchymal stem cells. IUBMB Life 2024; 76:1234-1251. [PMID: 39134088 PMCID: PMC11580377 DOI: 10.1002/iub.2905] [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: 02/05/2024] [Accepted: 06/25/2024] [Indexed: 11/22/2024]
Abstract
Glioblastoma (GB) is a lethal brain tumor that rapidly adapts to the dynamic changes of the tumor microenvironment (TME). Mesenchymal stem/stromal cells (MSCs) are one of the stromal components of the TME playing multiple roles in tumor progression. GB progression is prompted by the immunosuppressive microenvironment characterized by high concentrations of the nucleoside adenosine (ADO). ADO acts as a signaling molecule through adenosine receptors (ARs) but also as a genetic and metabolic regulator. Herein, the effects of high extracellular ADO concentrations were investigated in a human glioblastoma cellular model (U343MG) and MSCs. The modulation of the purinome machinery, i.e., the ADO production (CD39, CD73, and adenosine kinase [ADK]), transport (equilibrative nucleoside transporters 1 (ENT1) and 2 (ENT2)), and degradation (adenosine deaminase [ADA]) were investigated in both cell lines to evaluate if ADO could affect its cell management in a positive or negative feed-back loop. Results evidenced a different behavior of GB and MSC cells upon exposure to high extracellular ADO levels: U343MG were less sensitive to the ADO concentration and only a slight increase in ADK and ENT1 was evidenced. Conversely, in MSCs, the high extracellular ADO levels reduced the ADK, ENT1, and ENT2 expression, which further sustained the increase of extracellular ADO. Of note, MSCs primed with the GB-conditioned medium or co-cultured with U343MG cells were not affected by the increase of extracellular ADO. These results evidenced how long exposure to ADO could produce different effects on cancer cells with respect to MSCs, revealing a negative feedback loop that can support the GB immunosuppressive microenvironment. These results improve the knowledge of the ADO role in the maintenance of TME, which should be considered in the development of therapeutic strategies targeting adenosine pathways as well as cell-based strategies using MSCs.
Collapse
Affiliation(s)
| | - Lara Russo
- Department of PharmacyUniversity of PisaPisaItaly
| | | | | | | | | | | |
Collapse
|
3
|
Zulaziz N, Chai SJ, Lim KP. The origins, roles and therapies of cancer associated fibroblast in liver cancer. Front Oncol 2023; 13:1151373. [PMID: 37035187 PMCID: PMC10076538 DOI: 10.3389/fonc.2023.1151373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/03/2023] [Indexed: 04/11/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of liver cancer. It is often preceded by chronic inflammation such as liver fibrosis and cirrhosis. Different cell types are believed to give rise to liver-specific cancer associated fibroblast (CAF), these include resident fibroblast, hepatic stellate cell, liver cancer cell, hepatic sinusoidal endothelial cell and mesenchymal stromal cell. The abundance of fibroblasts has contributed to the cancer progression, immune modulation and treatment resistance in HCC. In this review, we discussed the origins, subtypes and roles of cancer associated fibroblasts in HCC. Their specific roles in shaping the tumor microenvironment, facilitating cancer growth, and modulating different immune cell types to confer a permissive environment for cancer growth. CAF is now an attractive therapeutic target for cancer treatment, however specific therapeutic development in HCC is still lacking. Hence, we have included preclinical and clinical development of CAF-specific interventions for other cancer types in this review. However, most CAF-specific therapies have resulted in disappointing clinical outcomes, likely due to the difficulties in differentiating CAF from normal fibroblast. A thorough understanding of the characteristics and functionalities of CAF is warranted to further improve the therapeutic efficacy of anti-CAF therapies.
Collapse
|
4
|
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: 16] [Impact Index Per Article: 5.3] [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
|
5
|
Zhang Z, Zhou X, Guo J, Zhang F, Qian Y, Wang G, Duan M, Wang Y, Zhao H, Yang Z, Liu Z, Jiang X. TA-MSCs, TA-MSCs-EVs, MIF: their crosstalk in immunosuppressive tumor microenvironment. J Transl Med 2022; 20:320. [PMID: 35842634 PMCID: PMC9287873 DOI: 10.1186/s12967-022-03528-y] [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: 04/06/2022] [Accepted: 07/08/2022] [Indexed: 11/24/2022] Open
Abstract
As an important component of the immunosuppressive tumor microenvironment (TME), it has been established that mesenchymal stem cells (MSCs) promote the progression of tumor cells. MSCs can directly promote the proliferation, migration, and invasion of tumor cells via cytokines and chemokines, as well as promote tumor progression by regulating the functions of anti-tumor immune and immunosuppressive cells. MSCs-derived extracellular vesicles (MSCs-EVs) contain part of the plasma membrane and signaling factors from MSCs; therefore, they display similar effects on tumors in the immunosuppressive TME. The tumor-promoting role of macrophage migration inhibitory factor (MIF) in the immunosuppressive TME has also been revealed. Interestingly, MIF exerts similar effects to those of MSCs in the immunosuppressive TME. In this review, we summarized the main effects and related mechanisms of tumor-associated MSCs (TA-MSCs), TA-MSCs-EVs, and MIF on tumors, and described their relationships. On this basis, we hypothesized that TA-MSCs-EVs, the MIF axis, and TA-MSCs form a positive feedback loop with tumor cells, influencing the occurrence and development of tumors. The functions of these three factors in the TME may undergo dynamic changes with tumor growth and continuously affect tumor development. This provides a new idea for the targeted treatment of tumors with EVs carrying MIF inhibitors.
Collapse
Affiliation(s)
- Zhenghou Zhang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiangyu Zhou
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jinshuai Guo
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fusheng Zhang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yiping Qian
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Guang Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Meiqi Duan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yutian Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Haiying Zhao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhi Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zunpeng Liu
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| | - Xiaofeng Jiang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
6
|
Mesenchymal Stem Cell-Based Therapy as a New Approach for the Treatment of Systemic Sclerosis. Clin Rev Allergy Immunol 2022; 64:284-320. [PMID: 35031958 DOI: 10.1007/s12016-021-08892-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 12/13/2022]
Abstract
Systemic sclerosis (SSc) is an intractable autoimmune disease with unmet medical needs. Conventional immunosuppressive therapies have modest efficacy and obvious side effects. Targeted therapies with small molecules and antibodies remain under investigation in small pilot studies. The major breakthrough was the development of autologous haematopoietic stem cell transplantation (AHSCT) to treat refractory SSc with rapidly progressive internal organ involvement. However, AHSCT is contraindicated in patients with advanced visceral involvement. Mesenchymal stem cells (MSCs) which are characterized by immunosuppressive, antifibrotic and proangiogenic capabilities may be a promising alternative option for the treatment of SSc. Multiple preclinical and clinical studies on the use of MSCs to treat SSc are underway. However, there are several unresolved limitations and safety concerns of MSC transplantation, such as immune rejections and risks of tumour formation, respectively. Since the major therapeutic potential of MSCs has been ascribed to their paracrine signalling, the use of MSC-derived extracellular vesicles (EVs)/secretomes/exosomes as a "cell-free" therapy might be an alternative option to circumvent the limitations of MSC-based therapies. In the present review, we overview the current knowledge regarding the therapeutic efficacy of MSCs in SSc, focusing on progresses reported in preclinical and clinical studies using MSCs, as well as challenges and future directions of MSC transplantation as a treatment option for patients with SSc.
Collapse
|
7
|
Rat Adipose-Derived Stromal Cells (ADSCs) Increases the Glioblastoma Growth and Decreases the Animal Survival. Stem Cell Rev Rep 2021; 18:1495-1509. [PMID: 34403074 DOI: 10.1007/s12015-021-10227-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2021] [Indexed: 12/22/2022]
Abstract
Many studies have shown that mesenchymal stromal cells (MSCs) and their secreted factors may modulate the biology of tumor cells. However, how these interactions happen in vivo remains unclear. In the present study, we investigated the effects of rat adipose-derived stromal cells (ADSCs) and their conditioned medium (ADSC-CM) in glioma tumor growth and malignancy in vivo. Our results showed that when we co-injected C6 cells plus ADSCs into the rat brains, the tumors generated were larger and the animals exhibited shorter survival, when compared with tumors of the animals that received only C6 cells or C6 cells pre-treated with ADSC-CM. We further showed that the animals that received C6 plus ADSC did not present enhanced expression of CD73 (a gene highly expressed in ADSCs), indicating that the tumor volume observed in these animals was not a mere consequence of the higher density of cells administered in this group. Finally, we showed that the animals that received C6 + ADSC presented tumors with larger necrosis areas and greater infiltration of immune cells. These results indicate that the immunoregulatory properties of ADSCs and its contribution to tumor stroma can support tumor growth leading to larger zones of necrosis, recruitment of immune cells, thus facilitating tumor progression. Our data provide new insights into the way by which ADSCs and tumor cells interact and highlight the importance of understanding the fate and roles of MSCs in tumor sites in vivo, as well as their intricate crosstalk with cancer cells.
Collapse
|
8
|
Zhang Y, Zhu J, Xu H, Yi Q, Yan L, Ye L, Zhang X, Xie M, Tan B. Time-Dependent Internalization of S100B by Mesenchymal Stem Cells via the Pathways of Clathrin- and Lipid Raft-Mediated Endocytosis. Front Cell Dev Biol 2021; 9:674995. [PMID: 34381770 PMCID: PMC8351554 DOI: 10.3389/fcell.2021.674995] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/06/2021] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are promising tools for cancer therapy, but there is a risk of malignant transformation in their clinical application. Our previous work revealed that the paracrine protein S100B in the glioma microenvironment induces malignant transformation of MSCs and upregulates intracellular S100B, which could affect cell homeostasis by interfering with p53. The purpose of this study was to investigate whether extracellular S100B can be internalized by MSCs and the specific endocytic pathway involved in S100B internalization. By using real-time confocal microscopy and structured illumination microscopy (SIM), we visualized the uptake of fluorescently labeled S100B protein (S100B-Alexa488) and monitored the intracellular trafficking of internalized vesicles. The results showed that S100B-Alexa488 was efficiently internalized into MSCs in a time-dependent manner and transported through endolysosomal pathways. After that, we used chemical inhibitors and RNA interference approaches to investigate possible mechanisms involved in S100B-Alexa488 uptake. The internalization of S100B-Alexa488 was inhibited by pitstop-2 or dyngo-4a treatment or RNA-mediated silencing of clathrin or dynamin, and the lipid raft-mediated endocytosis inhibitors nystatin and MβCD. In conclusion, our findings show that clathrin and lipid rafts contribute to the internalization of S100B-Alexa488, which provides promising interventions for the safe application of MSCs in glioma therapy.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jing Zhu
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Hao Xu
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
- Department of Clinical Laboratory, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Yi
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Liang Yan
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Liang Ye
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xinyuan Zhang
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Min Xie
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Bin Tan
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
9
|
Sun J, Huang J, Bao G, Zheng H, Wang C, Wei J, Fu Y, Qiu J, Liao Y, Cai J. MRI detection of the malignant transformation of stem cells through reporter gene expression driven by a tumor-specific promoter. Stem Cell Res Ther 2021; 12:284. [PMID: 33980305 PMCID: PMC8117323 DOI: 10.1186/s13287-021-02359-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 04/27/2021] [Indexed: 01/10/2023] Open
Abstract
Background Existing evidence has shown that mesenchymal stem cells (MSCs) can undergo malignant transformation, which is a serious limitation of MSC-based therapies. Therefore, it is necessary to monitor malignant transformation of MSCs via a noninvasive imaging method. Although reporter gene-based magnetic resonance imaging (MRI) has been successfully applied to longitudinally monitor MSCs, this technique cannot distinguish the cells before and after malignant transformation. Herein, we investigated the feasibility of using a tumor-specific promoter to drive reporter gene expression for MRI detection of the malignant transformation of MSCs. Methods The reporter gene ferritin heavy chain (FTH1) was modified by adding a promoter from the tumor-specific gene progression elevated gene-3 (PEG3) and transduced into MSCs to obtain MSCs-PEG3-FTH1. Cells were induced to undergo malignant transformation via indirect coculture with C6 glioma cells, and these transformed cells were named MTMSCs-PEG3-FTH1. Western blot analysis of FTH1 expression, Prussian blue staining and transmission electron microscopy (TEM) to detect intracellular iron, and MRI to detect signal changes were performed before and after malignant transformation. Then, the cells before and after malignant transformation were inoculated subcutaneously into nude mice, and MRI was performed to observe the signal changes in the xenografts. Results After induction of malignant transformation, MTMSCs demonstrated tumor-like features in morphology, proliferation, migration, and invasion. FTH1 expression was significantly increased in MTMSCs-PEG3-FTH1 compared with MSCs-PEG3-FTH1. Prussian blue staining and TEM showed a large amount of iron particles in MTMSCs-PEG3-FTH1 but a minimal amount in MSCs-PEG3-FTH1. MRI demonstrated that the T2 value was significantly decreased in MTMSCs-PEG3-FTH1 compared with MSCs-PEG3-FTH1. In vivo, mass formation was observed in the MTMSCs-PEG3-FTH1 group but not the MSCs-PEG3-FTH1 group. T2-weighted MRI showed a significant signal decrease, which was correlated with iron accumulation in the tissue mass. Conclusions We developed a novel MRI model based on FTH1 reporter gene expression driven by the tumor-specific PEG3 promoter. This approach could be applied to sensitively detect the occurrence of MSC malignant transformation.
Collapse
Affiliation(s)
- Jun Sun
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China.,Department of Radiology, Chongqing University Central Hospital, Chongqing, 400014, China
| | - Jie Huang
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China
| | - Guangcheng Bao
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China
| | - Helin Zheng
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Cui Wang
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Jie Wei
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China
| | - Yuanqiao Fu
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China
| | - Jiawen Qiu
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China
| | - Yifan Liao
- Department of Nuclear Medicine, Xinqiao Hospital affiliated with Third Military Medical University, Chongqing, 400037, China
| | - Jinhua Cai
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China. .,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China. .,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China.
| |
Collapse
|
10
|
Wang C, Hu J, Chen Z, Wang Y, Lu S, Zhang Y, Li Y, Xiang Y, Ji Y, Zeng C, Ding Y, Wang W. Reversibility of hAT-MSCs phenotypic and metabolic changes after exposure to and withdrawal from HCC-conditioned medium through regulation of the ROS/MAPK/HIF-1α signaling pathway. Stem Cell Res Ther 2020; 11:506. [PMID: 33246501 PMCID: PMC7694319 DOI: 10.1186/s13287-020-02010-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 11/03/2020] [Indexed: 01/14/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) play an important role in tumor progression; concomitantly, MSCs also undergo profound changes in the tumor microenvironment (TME). These changes can directly impact the application and efficacy of MSC-based anti-tumor therapy. However, few studies have focused on the regulation of MSC fate in TME, which will limit the progress of MSC-based anti-tumor therapy. Herein, we investigated the effects of conditioned medium from human hepatocellular carcinoma cells (HCC-CM) on the phenotype and glucose metabolism of human adipose tissue-derived MSCs (hAT-MSCs). Methods The passage 2 (P2) to passage 3 (P3) hAT-MSCs were exposed to conditioned medium from Hep3B, Huh7 and HCCLM3 cells for 4–8 weeks in vitro. Then, immunofluorescent, CCK-8 assay, EdU assay, Transwell assay, and flow cytometry were used to assess the alterations in cell phenotype in terms of cell morphology, secretory profiles, proliferation, migration, invasion, cell cycle, and apoptosis. In addition, glucose metabolism was evaluated by related kits. Next, the treated hAT-MSCs were subjected to withdrawal from HCC-CM for 2–4 weeks, and alterations in phenotype and glucose metabolism were reevaluated. Finally, the molecular mechanism was clarified by Western blotting. Results The results revealed that after exposure to HCC-CM, hAT-MSCs developed a stellate-shaped morphology. In association with cytoskeleton remodeling, hAT-MSCs showed enhanced capacities for migration and invasion, while cell proliferation was inhibited by regulating the cell cycle by downregulating cyclins and cyclin-dependent kinases and activating the mitochondrial apoptosis pathway. In terms of glucose metabolism, our results showed mitochondrial dysfunction and elevated glycolysis of hAT-MSCs. However, interestingly, when the treated hAT-MSCs were subjected to withdrawal from HCC-CM, the alterations in phenotype and glucose metabolism could be reversed, but secretory phenotype and tumor-promoting properties appear to be permanent. Further studies showed that these changes in hAT-MSCs may be regulated by the ROS/MAPK/HIF-1α signaling pathway. Conclusion Taken together, the effects of long-term HCC-CM treatment on phenotype and glucose metabolism in hAT-MSCs are modest and largely reversible after withdrawal, but HCC-CM endow hAT-MSCs with permanent secretory phenotype and tumor-promoting properties. This is the first report on the reversal of phenotype and glucose metabolism in tumor-associated MSCs (TA-MSCs), it is anticipated that new insights into TA-MSCs will lead to the development of novel strategies for MSC-based anti-tumor therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-020-02010-0.
Collapse
Affiliation(s)
- Chenyang Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Jie Hu
- Department of Plastic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Zheng Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Yifan Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Sinan Lu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yuan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yufeng Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yucheng Xiang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yutian Ji
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Cheng Zeng
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Diseases of Zhejiang University, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Diseases of Zhejiang University, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
11
|
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: 0.6] [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
|
12
|
Wen L, Sun J, Chen X, Du R. miR-135b-dependent downregulation of S100B promotes neural stem cell differentiation in a hypoxia/ischemia-induced cerebral palsy rat model. Am J Physiol Cell Physiol 2020; 319:C955-C966. [PMID: 32491925 DOI: 10.1152/ajpcell.00481.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cerebral palsy (CP) is frequently caused by brain injury during pregnancy, delivery, or the immediate postnatal period. The differentiation potential of neural stem cell (NSC) makes them effective in restoring injured tissues and organs with minimal risks of side effects. In this study, we identified a novel microRNA-135b (miR-135b) in CP and investigated its functional role in mediating NSC differentiation. CP models were established in Wistar rats and validated with the Y-maze test. Gain- and loss-of-function experimentation was performed on CP rats. Then NSCs were isolated and the expression patterns of miR-135b and S100B were altered in NSCs. S100B exhibited high expression in the hippocampus tissues of CP models, which was targeted by miR-135b. miR-135b elevation or S100B silencing resulted in promoted NSC differentiation, alleviated brain injury, and inhibited NSC apoptosis in hippocampus tissues of CP rats. S100B downregulation targeted by miR-135b overexpression contributed to the inactivation of the signal transducer and activator of transcription-3 (STAT3) pathway, which promoted NSC differentiation and proliferation but inhibited NSC apoptosis. Our results highlight the suppressor role played by miR-135b in CP by inducing NSC differentiation via inactivation of S100B-dependent STAT3 pathway.
Collapse
Affiliation(s)
- Linbao Wen
- Department of Neurosurgery, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, People's Republic of China
| | - Jingwei Sun
- Department of Neurosurgery, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, People's Republic of China
| | - Xionggao Chen
- Department of Neurosurgery, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, People's Republic of China
| | - Ruili Du
- Department of Radiology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, People's Republic of China
| |
Collapse
|
13
|
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: 46] [Impact Index Per Article: 9.2] [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
|
14
|
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: 1.5] [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
|
15
|
Sacca PA, Mazza ON, Scorticati C, Vitagliano G, Casas G, Calvo JC. Human Periprostatic Adipose Tissue: Secretome from Patients With Prostate Cancer or Benign Prostate Hyperplasia. Cancer Genomics Proteomics 2019; 16:29-58. [PMID: 30587498 DOI: 10.21873/cgp.20110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/10/2018] [Accepted: 10/12/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND/AIM Periprostatic adipose tissue (PPAT) directs tumour behaviour. Microenvironment secretome provides information related to its biology. This study was performed to identify secreted proteins by PPAT, from both prostate cancer and benign prostate hyperplasia (BPH) patients. PATIENTS AND METHODS Liquid chromatography-mass spectrometry-based proteomic analysis was performed in PPAT-conditioned media (CM) from patients with prostate cancer (CMs-T) (stage T3: CM-T3, stage T2: CM-T2) or benign disease (CM-BPH). RESULTS The highest number and diversity of proteins was identified in CM-T3. Locomotion was the biological process mainly associated to CMs-T and reproduction to CM-T3. Immune responses were enriched in CMs-T. Extracellular matrix and structural proteins were associated to CMs-T. CM-T3 was enriched in proteins with catalytic activity and CM-T2 in proteins with defense/immunity activity. Metabolism and energy pathways were enriched in CM-T3 and those with immune system functions in CMs-T. Transport proteins were enriched in CM-T2 and CM-BPH. CONCLUSION Proteins and pathways reported in this study could be useful to distinguish stages of disease and may become targets for novel therapies.
Collapse
Affiliation(s)
- Paula Alejandra Sacca
- Institute of Biology and Experimental Medicine (IBYME), CONICET, Buenos Aires, Argentina
| | - Osvaldo Néstor Mazza
- Department of Urology, School of Medicine, University of Buenos Aires, Clínical Hospital "José de San Martín", Buenos Aires, Argentina
| | - Carlos Scorticati
- Department of Urology, School of Medicine, University of Buenos Aires, Clínical Hospital "José de San Martín", Buenos Aires, Argentina
| | | | - Gabriel Casas
- Department of Pathology, Deutsches Hospital, Buenos Aires, Argentina
| | - Juan Carlos Calvo
- Institute of Biology and Experimental Medicine (IBYME), CONICET, Buenos Aires, Argentina.,Department of Biological Chemistry, School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
16
|
Cui X, Jing X, Yi Q, Xiang Z, Tian J, Tan B, Zhu J. IL22 furthers malignant transformation of rat mesenchymal stem cells, possibly in association with IL22RA1/STAT3 signaling. Oncol Rep 2019; 41:2148-2158. [PMID: 30816520 PMCID: PMC6412447 DOI: 10.3892/or.2019.7007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/05/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) hold great promise as potential therapies for tumors through the delivery of various anticancer agents. However, exogenous tissue‑derived MSCs, such as those of bone marrow, have exhibited a tendency for malignant transformation in the tumor microenvironment. This issue remains controversial and is poorly understood. In the present study, the role of interleukin 22 (IL22)/IL22 receptor subunit α 1 (IL22RA1) and signal transducer and activator of transcription 3 (STAT3) signaling in the malignant transformation of MSCs was investigated. Following isolation of rat MSCs and their indirect co‑culture with C6 glioma cells, the transformed MSCs exhibited tumor cell characteristics. The Cancer Genome Atlas‑Glioblastoma Multiforme analysis revealed that primary and recurrent glioblastomas have increased IL22RA1 expression, compared with normal tissues, whereas the expression of IL22 was low in glioblastoma and normal tissues. mRNA and protein expression levels of IL22RA1 were significantly increased in the MSCs co‑cultured with C6 glioma cells. Furthermore, MSCs incubated with IL22 exhibited increased proliferation, migration and invasion. STAT3 demonstrated activation and nuclear translocation in the presence of IL22. Additionally, STAT3 small interfering RNA significantly inhibited the migration and invasion ability of MSCs, and the expression of the STAT3 downstream targets cyclin D1 and B‑cell lymphoma‑extra large under IL22 stimulation, indicating that IL22 also promoted MSC migration and invasion through STAT3 signaling. These data indicated that IL22 serves a critical role in the malignant transformation of rat MSCs, which is associated with an enhancement of the IL22RA1/STAT3 signaling pathway in the tumor microenvironment.
Collapse
Affiliation(s)
- Xiangrong Cui
- Ministry of Education Key Laboratory of Child Development and Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Xuan Jing
- Clinical Laboratory, Shanxi Province People's Hospital, Taiyuan, Shanxi 030001, P.R. China
| | - Qin Yi
- Ministry of Education Key Laboratory of Child Development and Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Zhongping Xiang
- Ministry of Education Key Laboratory of Child Development and Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Jie Tian
- Cardiovascular Department (Internal Medicine), Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Bin Tan
- Ministry of Education Key Laboratory of Child Development and Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Jing Zhu
- Ministry of Education Key Laboratory of Child Development and Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| |
Collapse
|
17
|
Băbţan AM, Ilea A, Boşca BA, Crişan M, Petrescu NB, Collino M, Sainz RM, Gerlach JQ, Câmpian RS. Advanced glycation end products as biomarkers in systemic diseases: premises and perspectives of salivary advanced glycation end products. Biomark Med 2019; 13:479-495. [PMID: 30968701 DOI: 10.2217/bmm-2018-0448] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Advanced glycation end products (AGEs) are glycated proteins associated with high dry temperature food processing, coloring and flavor modification of food products. Previous studies on diet-related disease support the role of the glycation products as biomarkers in local and general proinflammatory response. Exogenous and endogenous AGEs are involved in chronic low-level inflammation, which underlies the onset of metabolic syndrome influenced by food intake, there by demonstrating their implication in diet-related pathologies. Although studies have revealed a strong association between the accumulation of AGEs and the occurrence/worsening of metabolic diseases, their routine use for the diagnosis or monitoring of local and general disease has not yet been reported.
Collapse
Affiliation(s)
- Anida M Băbţan
- Department of Oral Rehabilitation, Oral Health & Dental Office Management, Faculty of Dentistry, ‘Iuliu Haţieganu’ University of Medicine & Pharmacy Cluj-Napoca, Romania, Victor Babe? Street, no 15, 400012, Romania
| | - Aranka Ilea
- Department of Oral Rehabilitation, Oral Health & Dental Office Management, Faculty of Dentistry, ‘Iuliu Haţieganu’ University of Medicine & Pharmacy Cluj-Napoca, Romania, Victor Babe? Street, no 15, 400012, Romania
| | - Bianca A Boşca
- Department of Histology, Faculty of Medicine, ‘Iuliu Haţieganu’ University of Medicine & Pharmacy Cluj-Napoca, Romania, Louis Pasteur Street, no 4, Cluj-Napoca, 400349, Romania
| | - Maria Crişan
- Department of Histology, Faculty of Medicine, ‘Iuliu Haţieganu’ University of Medicine & Pharmacy Cluj-Napoca, Romania, Louis Pasteur Street, no 4, Cluj-Napoca, 400349, Romania
| | - Nausica B Petrescu
- Department of Oral Rehabilitation, Oral Health & Dental Office Management, Faculty of Dentistry, ‘Iuliu Haţieganu’ University of Medicine & Pharmacy Cluj-Napoca, Romania, Victor Babe? Street, no 15, 400012, Romania
| | - Massimo Collino
- Department of Drug Science & Technology, University of Turin, Corso Raffaello 33, 10125 Torino, Italy
| | - Rosa M Sainz
- Department of Morphology & Cell Biology, University of Oviedo, Campus del Cristo. C/Julián Clavería 6. 33006 Oviedo, Spain
| | - Jared Q Gerlach
- Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland Galway, H91 CF50 Galway, Ireland
| | - Radu S Câmpian
- Department of Oral Rehabilitation, Oral Health & Dental Office Management, Faculty of Dentistry, ‘Iuliu Haţieganu’ University of Medicine & Pharmacy Cluj-Napoca, Romania, Victor Babe? Street, no 15, 400012, Romania
| |
Collapse
|
18
|
Ma Z, Cui X, Lu L, Chen G, Yang Y, Hu Y, Lu Y, Cao Z, Wang Y, Wang X. Exosomes from glioma cells induce a tumor-like phenotype in mesenchymal stem cells by activating glycolysis. Stem Cell Res Ther 2019; 10:60. [PMID: 30770778 PMCID: PMC6377719 DOI: 10.1186/s13287-019-1149-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/23/2018] [Accepted: 01/21/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Exosomes are nanoscale membrane vesicles secreted by both normal and cancer cells, and cancer cell-derived exosomes play an important role in the cross-talk between cancer cells and other cellular components in the tumor microenvironment. Mesenchymal stem cells (MSCs) have tropism for tumors and have been used as tumor-tropic vectors for tumor therapy; however, the safety of such therapeutic use of MSCs is unknown. In this study, we investigated the role of glioma cell-derived exosomes in the tumor-like phenotype transformation of human bone marrow mesenchymal stem cells (hBMSCs) and explored the underlying molecular mechanisms. METHODS The effect of exosomes from U251 glioma cells on the growth of hBMSCs was evaluated with the CCK-8 assay, KI67 staining, and a cell cycle distribution assessment. The migration and invasion of hBMSCs were evaluated with a Transwell assay. A proteomics and bioinformatics approach, together with Western blotting and reverse transcriptase-polymerase chain reaction, was used to investigate the effect of U251 cell-derived exosomes on the proteome of hBMSCs. RESULTS U251 cell-derived exosomes induced a tumor-like phenotype in hBMSCs by enhancing their proliferation, migration, and invasion and altering the production of proteins involved in the regulation of the cell cycle. Moreover, U251 cell-derived exosomes promoted the production of the metastasis-related proteins MMP-2 and MMP-9, glioma marker GFAP, and CSC markers (CD133 and Nestin). The ten differentially expressed proteins identified participated in several biological processes and exhibited various molecular functions, mainly related to the inactivation of glycolysis. Western blotting showed that U251 cell-derived exosomes upregulated the levels of Glut-1, HK-2, and PKM-2, leading to the induction of glucose consumption and generation of lactate and ATP. Treatment with 2-deoxy-D-glucose significantly reversed these effects of U251 cell-derived exosomes on hBMSCs. CONCLUSIONS Our data demonstrate that glioma cell-derived exosomes activate glycolysis in hBMSCs, resulting in their tumor-like phenotype transformation. This suggests that interfering with the interaction between exosomes and hBMSCs in the tumor microenvironment has potential as a therapeutic approach for glioma. ᅟ.
Collapse
Affiliation(s)
- Zhanjun Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000 Gansu China
| | - Xue Cui
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 Gansu China
| | - Li Lu
- Institute of Pharmacology, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000 Gansu China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou, 730000 Gansu China
- School of Basic Medical Sciences of Lanzhou University, School of Medicine, 205 Tianshui Rd South, Lanzhou, 730000 Gansu China
| | - Guohu Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000 Gansu China
| | - Yang Yang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000 Gansu China
| | - Yan Hu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000 Gansu China
| | - Yubao Lu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000 Gansu China
| | - Zhangqi Cao
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000 Gansu China
| | - Yan Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000 Gansu China
| | - Xuexi Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000 Gansu China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou, 730000 Gansu China
- School of Basic Medical Sciences of Lanzhou University, School of Medicine, 205 Tianshui Rd South, Lanzhou, 730000 Gansu China
| |
Collapse
|
19
|
Zhang Q, Xiang W, Yi DY, Xue BZ, Wen WW, Abdelmaksoud A, Xiong NX, Jiang XB, Zhao HY, Fu P. Current status and potential challenges of mesenchymal stem cell-based therapy for malignant gliomas. Stem Cell Res Ther 2018; 9:228. [PMID: 30143053 PMCID: PMC6109313 DOI: 10.1186/s13287-018-0977-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Glioma, which accounts for more than 30% of primary central nervous system tumours, is characterised by symptoms such as headaches, epilepsy, and blurred vision. Glioblastoma multiforme is the most aggressive, malignant, and lethal brain tumour in adults. Even with progressive combination treatment with surgery, radiotherapy, and chemotherapy, the prognosis for glioma patients is still extremely poor. Compared with the poor outcome and slowly developing technologies for surgery and radiotherapy, the application of targeted chemotherapy with a new mechanism has become a research focus in this field. Moreover, targeted therapy is promising for most solid tumours. The tumour-tropic ability of stem cells, including neural stem cells and mesenchymal stem cells, provides an alternative therapeutic approach. Thus, mesenchymal stem cell-based therapy is based on a tumour-selective capacity and has been thought to be an effective anti-tumour option over the past decades. An increasing number of basic studies on mesenchymal stem cell-based therapy for gliomas has yielded complex outcomes. In this review, we summarise the biological characteristics of human mesenchymal stem cells, and the current status and potential challenges of mesenchymal stem cell-based therapy in patients with malignant gliomas.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Ave. Jiefang No.1277, Wuhan, 430022, People's Republic of China
| | - Wei Xiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Ave. Jiefang No.1277, Wuhan, 430022, People's Republic of China
| | - Dong-Ye Yi
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Ave. Jiefang No.1277, Wuhan, 430022, People's Republic of China
| | - Bing-Zhou Xue
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Ave. Jiefang No.1277, Wuhan, 430022, People's Republic of China
| | - Wan-Wan Wen
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No. 2, Anzhen Road, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Ahmed Abdelmaksoud
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Ave. Jiefang No.1277, Wuhan, 430022, People's Republic of China
| | - Nan-Xiang Xiong
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Ave. Jiefang No.1277, Wuhan, 430022, People's Republic of China
| | - Xiao-Bing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Ave. Jiefang No.1277, Wuhan, 430022, People's Republic of China
| | - Hong-Yang Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Ave. Jiefang No.1277, Wuhan, 430022, People's Republic of China
| | - Peng Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Ave. Jiefang No.1277, Wuhan, 430022, People's Republic of China.
| |
Collapse
|
20
|
Rosmaninho P, Mükusch S, Piscopo V, Teixeira V, Raposo AA, Warta R, Bennewitz R, Tang Y, Herold-Mende C, Stifani S, Momma S, Castro DS. Zeb1 potentiates genome-wide gene transcription with Lef1 to promote glioblastoma cell invasion. EMBO J 2018; 37:e97115. [PMID: 29903919 PMCID: PMC6068449 DOI: 10.15252/embj.201797115] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 04/27/2018] [Accepted: 05/07/2018] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma is the most common and aggressive brain tumor, with a subpopulation of stem-like cells thought to mediate its recurring behavior and therapeutic resistance. The epithelial-mesenchymal transition (EMT) inducing factor Zeb1 was linked to tumor initiation, invasion, and resistance to therapy in glioblastoma, but how Zeb1 functions at molecular level and what genes it regulates remain poorly understood. Contrary to the common view that EMT factors act as transcriptional repressors, here we show that genome-wide binding of Zeb1 associates with both activation and repression of gene expression in glioblastoma stem-like cells. Transcriptional repression requires direct DNA binding of Zeb1, while indirect recruitment to regulatory regions by the Wnt pathway effector Lef1 results in gene activation, independently of Wnt signaling. Amongst glioblastoma genes activated by Zeb1 are predicted mediators of tumor cell migration and invasion, including the guanine nucleotide exchange factor Prex1, whose elevated expression is predictive of shorter glioblastoma patient survival. Prex1 promotes invasiveness of glioblastoma cells in vivo highlighting the importance of Zeb1/Lef1 gene regulatory mechanisms in gliomagenesis.
Collapse
Affiliation(s)
- Pedro Rosmaninho
- Molecular Neurobiology Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Susanne Mükusch
- Institute of Neurology (Edinger Institute), Frankfurt University Medical School, Frankfurt, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Valerio Piscopo
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Vera Teixeira
- Molecular Neurobiology Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Alexandre Asf Raposo
- Molecular Neurobiology Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Rolf Warta
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Romina Bennewitz
- Institute of Neurology (Edinger Institute), Frankfurt University Medical School, Frankfurt, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yeman Tang
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Stefano Stifani
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Stefan Momma
- Institute of Neurology (Edinger Institute), Frankfurt University Medical School, Frankfurt, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Diogo S Castro
- Molecular Neurobiology Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| |
Collapse
|