1
|
Khaveh N, Buschow R, Metzger J. Deciphering transcriptome patterns in porcine mesenchymal stem cells promoting phenotypic maintenance and differentiation by key driver genes. Front Cell Dev Biol 2024; 12:1478757. [PMID: 39568509 PMCID: PMC11576426 DOI: 10.3389/fcell.2024.1478757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/28/2024] [Indexed: 11/22/2024] Open
Abstract
Mesenchymal stem cells (MSC) are fibroblast-like non-hematopoietic cells with self-renewal and differentiation capacity, and thereby great potential in regeneration and wound healing. MSC populations are heterogeneous not only inherently, but also among different model species. In particular, porcine MSC serve as a frequently used resource for translational research, due to pigs' distinctive closeness to human anatomy and physiology. However, information on gene expression profiles from porcine MSC and its dynamics during differentiation is sparse, especially with regard to cell surface and inner cell markers. In this study, we investigated the transcriptome of bone marrow-derived MSC and its differentiated cell types in a minipig breed for experimental research, known as Mini-LEWE, using bulk mRNA sequencing. Our data highlighted Rap1 signaling and downstream pathways PI3K-Akt and MAPK signaling as potential players for the maintenance of stemness of BM-MSC. In addition, we were able to link the process of differentiation to changes in the regulation of actin cytoskeleton. A total of 18 "BM-MSC differentiation driver markers" were identified, potentially promoting the process of differentiation into adipocytes, chondrocytes as well as osteocytes. Our results offer a new perspective on the molecular phenotype of porcine BM-MSC and the transcriptional responses in new differentiated progeny.
Collapse
Affiliation(s)
- Nadia Khaveh
- Institute of Animal Genomics, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Group Veterinary Functional Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - René Buschow
- Microscopy and Cryo-Electron Microscopy Facility, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Julia Metzger
- Institute of Animal Genomics, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Group Veterinary Functional Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| |
Collapse
|
2
|
Ren R, Chen Y, Zhou Y, Shen L, Chen Y, Lei J, Wang J, Liu X, Zhang N, Zhou D, Zhao H, Li Y. STIM1 promotes acquired resistance to sorafenib by attenuating ferroptosis in hepatocellular carcinoma. Genes Dis 2024; 11:101281. [PMID: 39281833 PMCID: PMC11402164 DOI: 10.1016/j.gendis.2024.101281] [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: 12/11/2023] [Revised: 02/08/2024] [Accepted: 03/03/2024] [Indexed: 09/18/2024] Open
Abstract
Dysregulated calcium (Ca2+) signaling pathways are associated with tumor cell death and drug resistance. In non-excitable cells, such as hepatocellular carcinoma (HCC) cells, the primary pathway for Ca2+ influx is through stromal interaction molecule 1 (STIM1)-mediated store-operated calcium entry (SOCE). Previous studies have demonstrated the involvement of STIM1-mediated SOCE in processes such as genesis, metastasis, and stem cell self-renewal of HCC. However, it remains unclear whether STIM1-mediated SOCE plays a role in developing acquired resistance to sorafenib in HCC patients. In this study, we established acquired sorafenib-resistant (SR) HCC cell lines by intermittently exposing them to increasing concentrations of sorafenib. Our results showed higher levels of STIM1 and stronger SOCE in SR cells compared with parental cells. Deleting STIM1 significantly enhanced sensitivity to sorafenib in SR cells, while overexpressing STIM1 promoted SR by activating SOCE. Mechanistically, STIM1 increased the transcription of SLC7A11 through the SOCE-CaN-NFAT pathway. Subsequently, up-regulated SLC7A11 increased glutathione synthesis, resulting in ferroptosis insensitivity and SR. Furthermore, combining the SOCE inhibitor SKF96365 with sorafenib significantly improved the sensitivity of SR cells to sorafenib both in vitro and in vivo. These findings suggest a potential strategy to overcome acquired resistance to sorafenib in HCC cells.
Collapse
Affiliation(s)
- Ran Ren
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400044, China
| | - Yu Chen
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yu Zhou
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Luyao Shen
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yang Chen
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Juan Lei
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Jingchun Wang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Xudong Liu
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400044, China
| | - Nan Zhang
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400044, China
| | - Dongqin Zhou
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Huakan Zhao
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400044, China
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yongsheng Li
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400044, China
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
3
|
Xue T, Wang X, Pan X, Liu M, Xu F. PTX promotes breast cancer migration and invasion by recruiting ATF4 to upregulate FGF19. Cell Signal 2024; 122:111309. [PMID: 39053672 DOI: 10.1016/j.cellsig.2024.111309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 07/03/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Widely-spread among women, breast cancer is a malignancy with fatalities, and chemotherapy is a vital treatment option for it. Recent studies have underscored the potential of chemotherapeutic agents such as paclitaxel, adriamycin, cyclophosphamide, and gemcitabine, among others, in facilitating tumor metastasis, with paclitaxel being extensively researched in this context. The molecular mechanism of these genes and their potential relevance to breast cancer is noteworthy. METHOD Clinical tissue specimens were used to analyze the expression and clinical significance of FGF19 or P-FGFR4 in patients with breast cancer before and after chemotherapy. qRT-PCR, ELISA, immunofluorescence and Western blotting were used to detect the expression level of FGF19 in breast cancer cells. The biological impacts of paclitaxel, FGF19, and ATF4 on breast cancer cells were assessed through CCK8, Transwell, and Western blot assays. The expression of ATF4 in breast cancer cells was determined through database analysis, Western blot analysis, qRT-PCR, and immunofluorescence. The direct interaction between FGF19 and ATF4 was confirmed by a luciferase assay, and Western blotting was used to assess the levels of key proteins in the stress response pathway. To confirm the effects of PTX and FGF19 in vivo, we established a lung metastasis model in nude mice. RESULTS FGF19 expression was increased in breast cancer patients after chemotherapy. Paclitaxel can boost the migration and invasion of breast cancer cells, accompanied by an increase in FGF19 expression. ATF4 might be involved in facilitating the enhancing effect of FGF19 on breast cancer cell migration. Finally, stimulation during paclitaxel treatment could trigger a stress response, influencing the expression of FGF19 and the migration of breast cancer cells. CONCLUSION These data suggest that paclitaxel regulates FGF19 expression through ATF4 and thus promotes breast cancer cell migration and invasion.
Collapse
Affiliation(s)
- Ting Xue
- School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China
| | - Xuezhen Wang
- School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China
| | - Xianjun Pan
- Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Mei Liu
- Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Faliang Xu
- School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China; Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China.
| |
Collapse
|
4
|
Shen L, Wang C, Ren R, Liu X, Zhou D, Chen Y, Zhou Y, Lei J, Xiao Y, Zhang N, Zhao H, Li Y. Fibroblast growth factor receptor 4 deficiency in macrophages aggravates experimental colitis by promoting M1-polarization. Inflamm Res 2024; 73:1493-1510. [PMID: 38981913 DOI: 10.1007/s00011-024-01910-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/13/2024] [Accepted: 06/27/2024] [Indexed: 07/11/2024] Open
Abstract
OBJECTIVE AND DESIGN Compelling evidence indicates that dysregulated macrophages may play a key role in driving inflammation in inflammatory bowel disease (IBD). Fibroblast growth factor (FGF)-19, which is secreted by ileal enterocytes in response to bile acids, has been found to be significantly lower in IBD patients compared to healthy individuals, and is negatively correlated with the severity of diarrhea. This study aims to explore the potential impact of FGF19 signaling on macrophage polarization and its involvement in the pathogenesis of IBD. METHODS The dextran sulfate sodium (DSS)-induced mouse colitis model was utilized to replicate the pathology of human IBD. Mice were created with a conditional knockout of FGFR4 (a specific receptor of FGF19) in myeloid cells, as well as mice that overexpressing FGF19 specifically in the liver. The severity of colitis was measured using the disease activity index (DAI) and histopathological staining. Various techniques such as Western Blotting, quantitative PCR, flow cytometry, and ELISA were employed to assess polarization and the expression of inflammatory genes. RESULTS Myeloid-specific FGFR4 deficiency exacerbated colitis in the DSS mouse model. Deletion or inhibition of FGFR4 in bone marrow-derived macrophages (BMDMs) skewed macrophages towards M1 polarization. Analysis of transcriptome sequencing data revealed that FGFR4 deletion in macrophages significantly increased the activity of the complement pathway, leading to an enhanced inflammatory response triggered by LPS. Mechanistically, FGFR4-knockout in macrophages promoted complement activation and inflammatory response by upregulating the nuclear factor-κB (NF-κB)-pentraxin3 (PTX3) pathway. Additionally, FGF19 suppressed these pathways and reduced inflammatory response by activating FGFR4 in inflammatory macrophages. Liver-specific overexpression of FGF19 also mitigated inflammatory responses induced by DSS in vivo. CONCLUSION Our study highlights the significance of FGF19-FGFR4 signaling in macrophage polarization and the pathogenesis of IBD, offering a potential new therapeutic target for IBD.
Collapse
Affiliation(s)
- Luyao Shen
- The Second Affiliated Hospital & Yuying Children's Hospital, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325030, Zhejiang, China
| | - Cong Wang
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325030, Zhejiang, China
| | - Ran Ren
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400044, China
| | - Xudong Liu
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400044, China
| | - Dongqin Zhou
- The Second Affiliated Hospital & Yuying Children's Hospital, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325030, Zhejiang, China
| | - Yu Chen
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yu Zhou
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Juan Lei
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yang Xiao
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400044, China
| | - Nan Zhang
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400044, China
| | - Huakan Zhao
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400044, China.
| | - Yongsheng Li
- The Second Affiliated Hospital & Yuying Children's Hospital, The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325030, Zhejiang, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
5
|
Chen T, Wang Q, Liu C, Zhang F, Bai Y, Jiao Y, Wang M, Bao S, Liu B, Shao M, Ma S, Ding Y. Ovatodiolide inhibited hepatocellular carcinoma stemness through SP1/MTDH/STAT3 signaling pathway. Chem Biol Interact 2024; 400:111161. [PMID: 39053793 DOI: 10.1016/j.cbi.2024.111161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/12/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Hepatocellular carcinoma (HCC) is characterized with high recurrence and mortality, and the clinical treatments for HCC are very limited. Hepatocellular carcinoma stem cells are the root of HCC progress, recurrence, and multidrug resistance. Ovatodiolide (OVA) is a bioactive diterpenoid served as an inflammatory and immunotherapeutic responses modulator. In this research, we found OVA inhibited HCC stemness through inhibiting MTDH gene transcription. Moreover, we firstly discovered transcription factor SP1 bound to the promoter region of MTDH to transcriptionally regulate MTDH level. Mechanically, we demonstrated OVA decreased SP1 protein stability to transcriptionally inhibit MTDH gene, and inhibited the nuclear translocation of p65, and then diminished IL-6 level to suppress JAK/STAT3 signaling pathway, eventually decreases CD133 level and the stemness of HCC. Furthermore, we demonstrated ACT004, OVA derivative with high metabolic stability towards cytochrome P450 enzymes, showed no genotoxicity and no accumulative or delayed toxicities after long-term administration in rats. And the in vivo efficacy experiments indicated ACT004 inhibited tumor growth of hepatocellular carcinoma. In conclusion, we revealed the mechanism of OVA in regulating HCC stemness, detected the toxicity of OVA derivative and evaluated the in vivo efficacy which lays a foundation for further discovery of anti-HCC stem cell agents and provide a new strategy for the application of OVA in clinical treatment.
Collapse
Affiliation(s)
- Tianyang Chen
- College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| | - Qin Wang
- College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| | - Can Liu
- College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| | - Fengyuan Zhang
- College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| | - Yongping Bai
- College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| | - Yan Jiao
- College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| | | | - Shiqi Bao
- Accendatech Co., Ltd., Tianjin, 300384, China
| | - Baofeng Liu
- Shan Dong Xinbo Pharmaceutical R&D Co., Ltd., Shan Dong, 251500, China
| | - Mingxiang Shao
- Shan Dong Xinbo Pharmaceutical R&D Co., Ltd., Shan Dong, 251500, China
| | - Shuoqian Ma
- College of Chemistry, Nankai University, Tianjin, 300071, China.
| | - Yahui Ding
- College of Chemistry, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
6
|
Zhu Z, Hu B, Zhu D, Li X, Chen D, Wu N, Rao Q, Zhang Z, Wang H, Zhu Y. Bromocriptine sensitivity in bromocriptine-induced drug-resistant prolactinomas is restored by inhibiting FGF19/FGFR4/PRL. J Endocrinol Invest 2024:10.1007/s40618-024-02408-0. [PMID: 38926262 DOI: 10.1007/s40618-024-02408-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
PURPOSE At present, various treatment strategies are available for pituitary adenomas, including medications, surgery and radiation. The guidelines indicate that pharmacological treatments, such as bromocriptine (BRC) and cabergoline (CAB), are important treatments for prolactinomas, but drug resistance is an urgent problem that needs to be addressed. Therefore, exploring the mechanism of drug resistance in prolactinomas is beneficial for clinical treatment. METHODS In our research, BRC-induced drug-resistant cells were established. Previous RNA sequencing data and an online database were used for preliminary screening of resistance-related genes. Cell survival was determined by Cell Counting Kit-8 (CCK-8) assay, colony formation assays and flow cytometry. Quantitative real-time polymerase chain reaction (qRT‒PCR), western blotting, immunohistochemistry, immunofluorescence and Co-immunoprecipitation (Co-IP) were used to assess the molecular changes and regulation. The therapeutic efficacy of BRC and FGFR4 inhibitor fisogatinib (FISO) combination was evaluated in drug-resistant cells and xenograft tumors in nude mice. RESULTS Consistent with the preliminary results of RNA sequencing and database screening, fibroblast growth factor 19 (FGF19) expression was elevated in drug-resistant cells and tumor samples. With FGF19 silencing, drug-resistant cells exhibited increased sensitivity to BRC and decreased intracellular phosphorylated fibroblast growth factor receptor 4 (FGFR4) levels. After confirming that FGF19 binds to FGFR4 in prolactinoma cells, we found that FGF19/FGFR4 regulated prolactin (PRL) synthesis through the ERK1/2 and JNK signaling pathways. Regarding the effect of targeting FGF19/FGFR4 on BRC efficacy, FISO and BRC synergistically inhibited the growth of tumor cells, promoted apoptosis and reduced PRL levels. CONCLUSION Overall, our study revealed FGF19/FGFR4 as a new mechanism involved in the drug resistance of prolactinomas, and combination therapy targeting the pathway could be helpful for the treatment of BRC-induced drug-resistant prolactinomas.
Collapse
Affiliation(s)
- Z Zhu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - B Hu
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - D Zhu
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - X Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - D Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - N Wu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Q Rao
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Z Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - H Wang
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
| | - Y Zhu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
- Department of Histology and Embryology, School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
7
|
Shen L, Li Y, Zhao H. Fibroblast growth factor signaling in macrophage polarization: impact on health and diseases. Front Immunol 2024; 15:1390453. [PMID: 38962005 PMCID: PMC11219802 DOI: 10.3389/fimmu.2024.1390453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/29/2024] [Indexed: 07/05/2024] Open
Abstract
Fibroblast growth factors (FGFs) are a versatile family of peptide growth factors that are involved in various biological functions, including cell growth and differentiation, embryonic development, angiogenesis, and metabolism. Abnormal FGF/FGF receptor (FGFR) signaling has been implicated in the pathogenesis of multiple diseases such as cancer, metabolic diseases, and inflammatory diseases. It is worth noting that macrophage polarization, which involves distinct functional phenotypes, plays a crucial role in tissue repair, homeostasis maintenance, and immune responses. Recent evidence suggests that FGF/FGFR signaling closely participates in the polarization of macrophages, indicating that they could be potential targets for therapeutic manipulation of diseases associated with dysfunctional macrophages. In this article, we provide an overview of the structure, function, and downstream regulatory pathways of FGFs, as well as crosstalk between FGF signaling and macrophage polarization. Additionally, we summarize the potential application of harnessing FGF signaling to modulate macrophage polarization.
Collapse
Affiliation(s)
- Luyao Shen
- The Second Affiliated Hospital & Yuying Children’s Hospital/The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yongsheng Li
- The Second Affiliated Hospital & Yuying Children’s Hospital/The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Huakan Zhao
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
8
|
Wang L, Ma Y, Zhang S, Yang Y, Huang B. NFATc2 promotes lactate and M2 macrophage polarization through USP17 in lung adenocarcinoma. Anticancer Drugs 2024; 35:385-396. [PMID: 38386130 DOI: 10.1097/cad.0000000000001582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
It is well known that immune cells including macrophages within the tumor microenvironment play an essential role in tumor progression. Here, we studied how NFATc2 regulated macrophage properties in lung adenocarcinoma. Higher expression of NFATc2 was observed in the lung adenocarcinoma tissues than in the normal lung tissues. Positive relationships were found between NFATc2 and genes associated with hypoxia and glycolysis in lung adenocarcinoma from the TCGA dataset. According to single-cell sequence data, NFATc2 was closely associated with infiltrating immune cells and was related to macrophage polarization. As a transcription factor, NFATc2 binding to the USP17 promoter region, that enhanced cell migration and lactate level in lung adenocarcinoma cells, and M2 polarization in macrophages. Furthermore, the NFATc2 inhibitor suppressed lactate and M2 macrophage polarization induced by NFATc2 and USP17. In conclusion, NFATc2 promotes lactate level and M2 macrophage polarization by transcriptionally regulating USP17 in lung adenocarcinoma.
Collapse
Affiliation(s)
- Liang Wang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing
| | - Yuanyuan Ma
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing
| | - Shanyuan Zhang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing
| | - Yue Yang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing
| | - Bo Huang
- Departments of Thoracic Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
9
|
Xiao Z, Wang S, Chen J, Li Y, Jiang Y, Tin VP, Liu J, Hu H, Wong MP, Pan Y, Yam JWP. The Dual Role of the NFATc2/galectin-9 Axis in Modulating Tumor-Initiating Cell Phenotypes and Immune Suppression in Lung Adenocarcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306059. [PMID: 38528665 PMCID: PMC11132051 DOI: 10.1002/advs.202306059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 03/04/2024] [Indexed: 03/27/2024]
Abstract
Tumor-initiating cells (TICs) resilience and an immunosuppressive microenvironment are aggressive oncogenic phenotypes that contribute to unsatisfactory long-term outcomes in lung adenocarcinoma (LUAD) patients. The molecular mechanisms mediating the interaction between TICs and immune tolerance have not been elucidated. The role of Galectin-9 in oncogenesis and immunosuppressive microenvironment is still unknown. This study explored the potential role of galectin-9 in TIC regulation and immune modulation in LUAD. The results show that galectin-9 supports TIC properties in LUAD. Co-culture of patient-derived organoids and matched peripheral blood mononuclear cells showed that tumor-secreted galectin-9 suppressed T cell cytotoxicity and induced regulatory T cells (Tregs). Clinically, galectin-9 is upregulated in human LUAD. High expression of galectin-9 predicted poor recurrence-free survival and correlated with high levels of Treg infiltration. LGALS9, the gene encoding galectin-9, is found to be transcriptionally regulated by the nuclear factor of activated T cells 2 (NFATc2), a previously reported TIC regulator, via in silico prediction and luciferase reporter assays. Overall, the results suggest that the NFATc2/galectin-9 axis plays a dual role in TIC regulation and immune suppression.
Collapse
Affiliation(s)
- Zhi‐Jie Xiao
- Scientific Research CentreThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhen518000China
- Shenzhen Key Laboratory of Bone Tissue Repair and Translational ResearchThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhen518000China
- Department of PathologySchool of Clinical MedicineThe University of Hong KongHong Kong999077Hong Kong
| | - Si‐Qi Wang
- Department of PathologySchool of Clinical MedicineThe University of Hong KongHong Kong999077Hong Kong
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
| | - Jun‐Jiang Chen
- Department of PhysiologySchool of MedicineJinan UniversityGuangzhou510000China
| | - Yun Li
- Department of Thoracic SurgeryThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityShenzhenChina
| | - Yuchen Jiang
- Scientific Research CentreThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhen518000China
| | - Vicky Pui‐Chi Tin
- Department of PathologySchool of Clinical MedicineThe University of Hong KongHong Kong999077Hong Kong
| | - Jia Liu
- Scientific Research CentreThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhen518000China
| | - Huiyi Hu
- Scientific Research CentreThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhen518000China
| | - Maria Pik Wong
- Department of PathologySchool of Clinical MedicineThe University of Hong KongHong Kong999077Hong Kong
| | - Yihang Pan
- Scientific Research CentreThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhen518000China
| | - Judy Wai Ping Yam
- Department of PathologySchool of Clinical MedicineThe University of Hong KongHong Kong999077Hong Kong
| |
Collapse
|
10
|
Gao W, Zhou J, Huang J, Zhang Z, Chen W, Zhang R, Kang T, Liao D, Zhong L. Up-regulation of RAN by MYBL2 maintains osteosarcoma cancer stem-like cells population during heterogeneous tumor generation. Cancer Lett 2024; 586:216708. [PMID: 38336287 DOI: 10.1016/j.canlet.2024.216708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/23/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
Intratumor heterogeneity is one of the major features of cancers, leading to aggressive disease and treatment failure. Cancer stem-like cells (CSCs) are believed to give rise to the heterogeneous cell types within tumors. Hence, understanding the regulatory mechanism underlying the recurrence process of heterogeneous tumor by CSCs could facilitate the development of CSC-targeted therapies. Here, utilizing single-cell transcriptomics, we present the molecular profile of osteosarcoma CSCs-derived heterogeneous tumors consisting of CSC clusters, osteoprogenitor and differentiated cell types, such as pre-osteoblasts, osteoblasts and chondroblasts. Furthermore, by constructing the comprehensive map of modulated genes during CSCs self-renewal and differentiation, we identify RAN exhibiting specific peak expression in osteosarcoma CSCs clusters which is transcriptionally up-regulated by MYBL2. Functionality, MYBL2-RAN pathway promotes the CSCs self-renewal by enhancing the nuclear accumulation of MYC protein, which in turn boosts the overexpression of RAN as a positive feedback. Importantly, blockage of MYBL2-RAN pathway sensitizes CSCs to cisplatin treatment and synergistically enhanced the cisplatin-induced cytotoxicity. Both MYBL2 and RAN are highly expressed in clinical osteosarcoma tissues which indicate poor prognosis. Collectively, our study provides advanced insights into the regeneration process of heterogeneous tumor originating from CSCs and highlights the MYBL2-RAN pathway as a promising target for CSC-based therapy in osteosarcoma.
Collapse
Affiliation(s)
- Weijie Gao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China; State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, PR China
| | - Jing Zhou
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, School of Medicine, Hubei Polytechnic University, Huangshi, PR China
| | - Jintao Huang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, PR China
| | - Zhiguang Zhang
- Sun Yat-sen University School of Medicine, Shenzhen, PR China
| | - Wanqi Chen
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Center of Digestive Diseases, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, PR China
| | - Ruhua Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Tiebang Kang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Dan Liao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| | - Li Zhong
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Center of Digestive Diseases, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, PR China.
| |
Collapse
|
11
|
Ji R, Chang L, An C, Zhang J. Proton-sensing ion channels, GPCRs and calcium signaling regulated by them: implications for cancer. Front Cell Dev Biol 2024; 12:1326231. [PMID: 38505262 PMCID: PMC10949864 DOI: 10.3389/fcell.2024.1326231] [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: 10/25/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Extracellular acidification of tumors is common. Through proton-sensing ion channels or proton-sensing G protein-coupled receptors (GPCRs), tumor cells sense extracellular acidification to stimulate a variety of intracellular signaling pathways including the calcium signaling, which consequently exerts global impacts on tumor cells. Proton-sensing ion channels, and proton-sensing GPCRs have natural advantages as drug targets of anticancer therapy. However, they and the calcium signaling regulated by them attracted limited attention as potential targets of anticancer drugs. In the present review, we discuss the progress in studies on proton-sensing ion channels, and proton-sensing GPCRs, especially emphasizing the effects of calcium signaling activated by them on the characteristics of tumors, including proliferation, migration, invasion, metastasis, drug resistance, angiogenesis. In addition, we review the drugs targeting proton-sensing channels or GPCRs that are currently in clinical trials, as well as the relevant potential drugs for cancer treatments, and discuss their future prospects. The present review aims to elucidate the important role of proton-sensing ion channels, GPCRs and calcium signaling regulated by them in cancer initiation and development. This review will promote the development of drugs targeting proton-sensing channels or GPCRs for cancer treatments, effectively taking their unique advantage as anti-cancer drug targets.
Collapse
Affiliation(s)
- Renhui Ji
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
- Department of Pathophysiology, Basic Medicine College of Inner Mongolia Medical University, Hohhot, China
| | - Li Chang
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
- Department of Pathophysiology, Basic Medicine College of Inner Mongolia Medical University, Hohhot, China
| | - Caiyan An
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
| | - Junjing Zhang
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
| |
Collapse
|
12
|
Guo Y, Feng Y, Jiang F, Hu L, Shan T, Li H, Liao H, Bao H, Shi H, Si Y. Down-regulating nuclear factor of activated T cells 1 alleviates cognitive deficits in a mouse model of sepsis-associated encephalopathy, possibly by stimulating hippocampal neurogenesis. Brain Res 2024; 1826:148731. [PMID: 38154504 DOI: 10.1016/j.brainres.2023.148731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/23/2023] [Accepted: 12/14/2023] [Indexed: 12/30/2023]
Abstract
Sepsis-associated encephalopathy (SAE) is a common complication of sepsis, and has been associated with increased morbidity and mortality. Nuclear factor of activated T cells (NFATs) 1, a transcriptional factor that regulates T cell development, activation and differentiation, has been implicated in neuronal plasticity. Here we examined the potential role of NFAT1 in sepsis-associated encephalopathy in mice. Adult male C57BL/6J mice received intracerebroventricular injections of short interfering RNA against NFAT1 or sex-determining region Y-box 2 (SOX2), or a scrambled control siRNA prior to cecal ligation and perforation (CLP). A group of mice receiving sham surgery were included as an additional control. CLP increased escape latency and decreased the number of crossings into, and total time spent within, the target quadrant in the Morris water maze test. CLP also decreased the freezing time in context-dependent, but not context-independent, fear conditioning test. Knockdown of either NFAT1 or SOX2 attenuated these behavioral deficits. NFAT1 knockdown also attenuated CLP-induced upregulation of SOX2, increased the numbers of nestin-positive cells and newborn astrocytes, reduced the number of immature newborn neurons, and promoted the G1 to S transition of neural stem cells in hippocampus. These findings suggest that NFAT1 may contribute to sepsis-induced behavioral deficits, possibly by promoting SOX2 signaling and neurogenesis.
Collapse
Affiliation(s)
- Yaoyi Guo
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Yue Feng
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Fan Jiang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Liang Hu
- Department of Pharmacology, Nanjing Medical University, No. 101 Longmiandadao Road, Jiangning District, Nanjing, Jiangsu Province 211166, People's Republic of China
| | - Tao Shan
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Haojia Li
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Hongsen Liao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Hongguang Bao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Hongwei Shi
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China
| | - Yanna Si
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, Jiangsu Province 210006, People's Republic of China.
| |
Collapse
|
13
|
Wang J, Xie Q, Wu L, Zhou Y, Xu Y, Chen Y, Zhang J, Ren R, Yang S, Li Y, Zhao H. Stromal interaction molecule 1/microtubule-associated protein 1A/1B-light chain 3B complex induces metastasis of hepatocellular carcinoma by promoting autophagy. MedComm (Beijing) 2024; 5:e482. [PMID: 38344399 PMCID: PMC10857778 DOI: 10.1002/mco2.482] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 10/28/2024] Open
Abstract
Metastasis is the leading cause of death in hepatocellular carcinoma (HCC) patients, and autophagy plays a crucial role in this process by orchestrating epithelial-mesenchymal transition (EMT). Stromal interaction molecule 1 (STIM1), a central regulator of store-operated calcium entry (SOCE) in nonexcitable cells, is involved in the development and spread of HCC. However, the impact of STIM1 on autophagy regulation during HCC metastasis remains unclear. Here, we demonstrate that STIM1 is temporally regulated during autophagy-induced EMT in HCC cells, and knocking out (KO) STIM1 significantly reduces both autophagy and EMT. Interestingly, STIM1 enhances autophagy through both SOCE-dependent and independent pathways. Mechanistically, STIM1 directly interacts with microtubule-associated protein 1A/1B-light chain 3B (LC3B) to form a complex via the sterile-α motif (SAM) domain, which promotes autophagosome formation. Furthermore, deletion of the SAM domain of STIM1 abolishes its binding with LC3B, leading to a decrease in autophagy and EMT in HCC cells. These findings unveil a novel mechanism by which the STIM1/LC3B complex mediates autophagy and EMT in HCC cells, highlighting a potential target for preventing HCC metastasis.
Collapse
Affiliation(s)
- Jingchun Wang
- Department of GastroenterologySecond Affiliated HospitalArmy Medical UniversityChongqingChina
| | - Qichao Xie
- Department of OncologyThe Third Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Lei Wu
- Department of Medical OncologyChongqing University Cancer HospitalChongqingChina
| | - Yu Zhou
- Department of Medical OncologyChongqing University Cancer HospitalChongqingChina
| | - Yanquan Xu
- Clinical Medicine Research CenterSecond Affiliated HospitalArmy Medical UniversityChongqingChina
| | - Yu Chen
- Department of Medical OncologyChongqing University Cancer HospitalChongqingChina
| | - Jiangang Zhang
- Department of Medical OncologyChongqing University Cancer HospitalChongqingChina
| | - Ran Ren
- Department of Medical OncologyChongqing University Cancer HospitalChongqingChina
| | - Shiming Yang
- Department of GastroenterologySecond Affiliated HospitalArmy Medical UniversityChongqingChina
| | - Yongsheng Li
- Department of Medical OncologyChongqing University Cancer HospitalChongqingChina
| | - Huakan Zhao
- Department of Medical OncologyChongqing University Cancer HospitalChongqingChina
| |
Collapse
|
14
|
Hu J, Chen K, Hong F, Gao G, Dai X, Yin H. METTL3 facilitates stemness properties and tumorigenicity of cancer stem cells in hepatocellular carcinoma through the SOCS3/JAK2/STAT3 signaling pathway. Cancer Gene Ther 2024; 31:228-236. [PMID: 38030810 DOI: 10.1038/s41417-023-00697-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/29/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023]
Abstract
Liver cancer stem cells (LCSCs) contribute to tumor recurrence and cancer cell proliferation in patients with hepatocellular carcinoma (HCC). METTL3-catalyzed m6A modification is relevant to the cancer stem cell (CSC) phenotype, including LCSCs. LCSCs were isolated from MHCC-97H and HepG2 cells through flow cytometry. UALCAN data were used to analyze the expression of METTL3 in liver hepatocellular carcinoma (LIHC) tissues. Loss- and gain-of-function experiments were utilized to assess the biological effects of METTL3 and SOCS3 on the proliferation and stemness phenotypes in vitro and in vivo. The mechanisms underlying the impact of METTL3 were explored using qPCR, MeRIP-qPCR, dual-luciferase reporter, and western blot assays. METTL3 was significantly upregulated in LIHC tissues according to the UALCAN database. METTL3 was highly expressed in LIHC and was significantly correlated with individual cancer stage, tumor grade and lymph node metastasis. Patients with low METTL3 expression had a longer overall survival time based on the data from UALCAN. In addition, the level of METTL3 was enhanced in LCSCs and decreased in non-LCSCs compared to HCC cells. Moreover, overexpression of METTL3 stimulated the proliferation and stemness of LCSCs in vitro and in vivo, while loss of METTL3 impeded it. Bioinformatics analysis combined with validation experiments determined that m6A was modified by METTL3-targeting SOCS3 mRNA. METTL3 had side effects regarding the stability of SOCS3 mRNA. SOCS3 overexpression impaired and SOCS3 depletion facilitated the development of LCSCs via the JAK2/STAT3 pathway. Furthermore, METTL3 depletion suppressed proliferation and stemness in LCSCs, which was restored by SOCS3 knockdown or colivelin treatment. We discovered that METTL3 facilitated the stemness and tumorigenicity of LCSCs by modifying SOCS3 mRNA with m6A.
Collapse
Affiliation(s)
- Jingjing Hu
- Department of Ultrasonography, Ningbo No. 2 Hospital, 315010, Ningbo, Zhejiang Province, P. R. China
| | - Ke Chen
- Ningbo City College of Vocational Technology, 315100, Ningbo, Zhejiang Province, P. R. China
| | - Fangfang Hong
- Department of Ultrasonography, Ningbo No. 2 Hospital, 315010, Ningbo, Zhejiang Province, P. R. China
| | - Guosheng Gao
- Clinical Laboratory, Ningbo No. 2 Hospital, 315010, Ningbo, Zhejiang Province, P. R. China
| | - Xiaoyu Dai
- Department of Anorectal Surgery, Ningbo No. 2 Hospital, 315010, Ningbo, Zhejiang Province, P. R. China
| | - Hua Yin
- Department of Ultrasonography, Ningbo No. 2 Hospital, 315010, Ningbo, Zhejiang Province, P. R. China.
| |
Collapse
|
15
|
Song Y, Lau HCH, Zhang X, Yu J. Bile acids, gut microbiota, and therapeutic insights in hepatocellular carcinoma. Cancer Biol Med 2023; 21:j.issn.2095-3941.2023.0394. [PMID: 38148326 PMCID: PMC10884537 DOI: 10.20892/j.issn.2095-3941.2023.0394] [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: 10/09/2023] [Accepted: 11/28/2023] [Indexed: 12/28/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent and aggressive liver malignancy. The interplay between bile acids (BAs) and the gut microbiota has emerged as a critical factor in HCC development and progression. Under normal conditions, BA metabolism is tightly regulated through a bidirectional interplay between gut microorganisms and BAs. The gut microbiota plays a critical role in BA metabolism, and BAs are endogenous signaling molecules that help maintain liver and intestinal homeostasis. Of note, dysbiotic changes in the gut microbiota during pathogenesis and cancer development can disrupt BA homeostasis, thereby leading to liver inflammation and fibrosis, and ultimately contributing to HCC development. Therefore, understanding the intricate interplay between BAs and the gut microbiota is crucial for elucidating the mechanisms underlying hepatocarcinogenesis. In this review, we comprehensively explore the roles and functions of BA metabolism, with a focus on the interactions between BAs and gut microorganisms in HCC. Additionally, therapeutic strategies targeting BA metabolism and the gut microbiota are discussed, including the use of BA agonists/antagonists, probiotic/prebiotic and dietary interventions, fecal microbiota transplantation, and engineered bacteria. In summary, understanding the complex BA-microbiota crosstalk can provide valuable insights into HCC development and facilitate the development of innovative therapeutic approaches for liver malignancy.
Collapse
Affiliation(s)
- Yang Song
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
- Department of Gastroenterology, Zhongshan Hospital Xiamen University, Xiamen 361004, China
| | - Harry CH Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
16
|
Su H, Shu S, Tang W, Zheng C, Zhao L, Fan H. ETV4 facilitates angiogenesis in hepatocellular carcinoma by upregulating MMP14 expression. Biochem Biophys Res Commun 2023; 684:149137. [PMID: 37897911 DOI: 10.1016/j.bbrc.2023.149137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023]
Abstract
Abnormal vascularization plays a crucial role in cell proliferation, tumor invasion and metastasis of hepatocellular carcinoma (HCC). It has been reported that ETV4 functions as an oncogenic gene in driving the carcinogenesis and progression, and promoting invasion and metastasis of HCC. However, the function of ETV4 on angiogenesis in HCC remains unclear. In the current study, immunohistochemistry showed that knockdown of ETV4 reduced angiogenesis in HCC xenograft tumor tissues. In vitro, tube formation assay verified that ETV4 expression promoted angiogenesis through simulating the angiogenic environment in HCC cells. Transcriptome sequencing indicated that MMP14 was one of the differentially expressed genes enriched in angiogenesis process. Subsequently, it was confirmed that MMP14 was regulated by ETV4 at the transcription level in HCC cells, clinical tissue samples and online databases. Further, we demonstrated that MMP14 induced angiogenesis in ETV4-mediated HCC microenvironment. Collectively, this research further reveals the biological mechanism of ETV4 in promoting the migration and invasion of HCC, and provides novel mechanistic insights and strategic guidance for anti-angiogenic therapy in HCC.
Collapse
Affiliation(s)
- Hongmeng Su
- Department of Medical Genetics and Developmental Biology, School of Medicine, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, China.
| | - Shihui Shu
- School of Life Science and Technology, Southeast University, Nanjing, China.
| | - Wenqing Tang
- School of Life Science and Technology, Southeast University, Nanjing, China.
| | - Chuqian Zheng
- Department of Medical Genetics and Developmental Biology, School of Medicine, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, China.
| | - Luyu Zhao
- Department of Medical Genetics and Developmental Biology, School of Medicine, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, China.
| | - Hong Fan
- Department of Medical Genetics and Developmental Biology, School of Medicine, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, China.
| |
Collapse
|
17
|
Guo F, Yang X, Hu C, Li W, Han W. Network Pharmacology Combined with Machine Learning to Reveal the Action Mechanism of Licochalcone Intervention in Liver Cancer. Int J Mol Sci 2023; 24:15935. [PMID: 37958916 PMCID: PMC10649909 DOI: 10.3390/ijms242115935] [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: 09/22/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
There are reports indicating that licochalcones can inhibit the proliferation, migration, and invasion of cancer cells by promoting the expression of autophagy-related proteins, inhibiting the expression of cell cycle proteins and angiogenic factors, and regulating autophagy and apoptosis. This study aims to reveal the potential mechanisms of licochalcone A (LCA), licochalcone B (LCB), licochalcone C (LCC), licochalcone D (LCD), licochalcone E (LCE), licochalcone F (LCF), and licochalcone G (LCG) inhibition in liver cancer through computer-aided screening strategies. By using machine learning clustering analysis to search for other structurally similar components in licorice, quantitative calculations were conducted to collect the structural commonalities of these components related to liver cancer and to identify key residues involved in the interactions between small molecules and key target proteins. Our research results show that the seven licochalcones molecules interfere with the cancer signaling pathway via the NF-κB signaling pathway, PDL1 expression and PD1 checkpoint pathway in cancer, and others. Glypallichalcone, Echinatin, and 3,4,3',4'-Tetrahydroxy-2-methoxychalcone in licorice also have similar structures to the seven licochalcones, which may indicate their similar effects. We also identified the key residues (including ASN364, GLY365, TRP366, and TYR485) involved in the interactions between ten flavonoids and the key target protein (nitric oxide synthase 2). In summary, we provide valuable insights into the molecular mechanisms of the anticancer effects of licorice flavonoids, providing new ideas for the design of small molecules for liver cancer drugs.
Collapse
Affiliation(s)
| | | | | | - Wannan Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, 2699 Qianjin Street, Changchun 130012, China; (F.G.); (X.Y.); (C.H.)
| | - Weiwei Han
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, 2699 Qianjin Street, Changchun 130012, China; (F.G.); (X.Y.); (C.H.)
| |
Collapse
|
18
|
Seyama Y, Sudo K, Hirose S, Hamano Y, Yamada T, Hiroyama T, Sasaki R, Hirai MY, Hyodo I, Tsuchiya K, Nakamura Y. Identification of a gene set that maintains tumorigenicity of the hepatocellular carcinoma cell line Li-7. Hum Cell 2023; 36:2074-2086. [PMID: 37610679 PMCID: PMC10587214 DOI: 10.1007/s13577-023-00967-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/31/2023] [Indexed: 08/24/2023]
Abstract
The identification and development of therapeutic targets in cancer stem cells that lead to tumor development, recurrence, metastasis, and drug resistance is an important goal in cancer research. The hepatocellular carcinoma cell line Li-7 contains functionally different types of cells. Cells with tumor-forming activity are enriched in cancer stem cell-like CD13+CD166- cells and this cell population gradually decreases during culture in conventional culture medium (RPMI1640 containing 10% fetal bovine serum). When Li-7 cells are cultured in mTeSR1, a medium developed for human pluripotent stem cells, CD13+CD166- cells, and their tumorigenicity is maintained. Here, we sought to identify the mechanisms of tumorigenicity in this sub-population. We compared gene expression profiles of CD13+CD166- cells with other cell sub-populations and identified nine overexpressed genes (ENPP2, SCGN, FGFR4, MCOLN3, KCNJ16, SMIM22, SMIM24, SERPINH1, and TMPRSS2) in CD13+CD166- cells. After transfer from mTeSR1 to RPMI1640 containing 10% fetal bovine serum, the expression of these nine genes decreased in Li-7 cells and they lost tumorigenicity. In contrast, when these genes of Li-7 cells were forcibly expressed in cultures using RPMI1640 containing 10% fetal bovine serum, Li-7 cells maintained tumorigenicity. A metabolome analysis using capillary electrophoresis-mass spectrometry showed that two metabolic pathways, "Alanine, aspartate and glutamate metabolism" and "Arginine biosynthesis" were activated in cancer stem-cell-like cells. Our analyses here showed potential therapeutic target genes and metabolites for treatment of cancer stem cells in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yusuke Seyama
- Division of Gastroenterology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Cell Engineering Division, RIKEN BioResource Research Center, Tsukuba, Japan
| | - Kazuhiro Sudo
- Cell Engineering Division, RIKEN BioResource Research Center, Tsukuba, Japan.
| | - Suguru Hirose
- Division of Gastroenterology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Cell Engineering Division, RIKEN BioResource Research Center, Tsukuba, Japan
| | - Yukako Hamano
- Department of Gastroenterology, Hitachi General Hospital, Hitachi, Japan
| | - Takeshi Yamada
- Division of Gastroenterology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Division of Clinical Research and Regional Innovation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takashi Hiroyama
- Cell Engineering Division, RIKEN BioResource Research Center, Tsukuba, Japan
| | - Ryosuke Sasaki
- RIKEN Center for Sustainable Resource Science, Yokohama, Japan
| | | | - Ichinosuke Hyodo
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Kiichiro Tsuchiya
- Division of Gastroenterology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Research Center, Tsukuba, Japan.
| |
Collapse
|
19
|
Liu Q, Huang J, Yan W, Liu Z, Liu S, Fang W. FGFR families: biological functions and therapeutic interventions in tumors. MedComm (Beijing) 2023; 4:e367. [PMID: 37750089 PMCID: PMC10518040 DOI: 10.1002/mco2.367] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/28/2023] [Accepted: 08/11/2023] [Indexed: 09/27/2023] Open
Abstract
There are five fibroblast growth factor receptors (FGFRs), namely, FGFR1-FGFR5. When FGFR binds to its ligand, namely, fibroblast growth factor (FGF), it dimerizes and autophosphorylates, thereby activating several key downstream pathways that play an important role in normal physiology, such as the Ras/Raf/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase, phosphoinositide 3-kinase (PI3K)/AKT, phospholipase C gamma/diacylglycerol/protein kinase c, and signal transducer and activator of transcription pathways. Furthermore, as an oncogene, FGFR genetic alterations were found in 7.1% of tumors, and these alterations include gene amplification, gene mutations, gene fusions or rearrangements. Therefore, FGFR amplification, mutations, rearrangements, or fusions are considered as potential biomarkers of FGFR therapeutic response for tyrosine kinase inhibitors (TKIs). However, it is worth noting that with increased use, resistance to TKIs inevitably develops, such as the well-known gatekeeper mutations. Thus, overcoming the development of drug resistance becomes a serious problem. This review mainly outlines the FGFR family functions, related pathways, and therapeutic agents in tumors with the aim of obtaining better outcomes for cancer patients with FGFR changes. The information provided in this review may provide additional therapeutic ideas for tumor patients with FGFR abnormalities.
Collapse
Affiliation(s)
- Qing Liu
- Cancer CenterIntegrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouGuangdongChina
| | - Jiyu Huang
- Cancer CenterIntegrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouGuangdongChina
| | - Weiwei Yan
- Cancer CenterIntegrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouGuangdongChina
| | - Zhen Liu
- Cancer CenterIntegrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouGuangdongChina
- Key Laboratory of Protein Modification and DegradationBasic School of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Shu Liu
- Department of Breast SurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
| | - Weiyi Fang
- Cancer CenterIntegrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
20
|
Lin Y, Song Y, Zhang Y, Shi M, Hou A, Han S. NFAT signaling dysregulation in cancer: Emerging roles in cancer stem cells. Biomed Pharmacother 2023; 165:115167. [PMID: 37454598 DOI: 10.1016/j.biopha.2023.115167] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023] Open
Abstract
The nuclear factor of activated T cells (NFAT) was first identified as a transcriptional regulator of activated T cells. The NFAT family is involved in the development of tumors. Furthermore, recent evidence reveals that NFAT proteins regulate the development of inflammatory and immune responses. New discoveries have also been made about the mechanisms by which NFAT regulates cancer progression through cancer stem cells (CSC). Here, we discuss the role of the NFAT family in the immune system and various cancer types.
Collapse
Affiliation(s)
- Yibin Lin
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yifu Song
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yaochuan Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Mengwu Shi
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ana Hou
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110001, China.
| | - Sheng Han
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
21
|
Wang R, Wang F, Lu S, Gao B, Kan Y, Yuan T, Xu Y, Yuan C, Guo D, Fu W, Yu X, Si Y. Adipose-derived stem cell/FGF19-loaded microfluidic hydrogel microspheres for synergistic restoration of critical ischemic limb. Bioact Mater 2023; 27:394-408. [PMID: 37122899 PMCID: PMC10131126 DOI: 10.1016/j.bioactmat.2023.04.006] [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: 10/08/2022] [Revised: 03/19/2023] [Accepted: 04/04/2023] [Indexed: 05/02/2023] Open
Abstract
The efficacy of stem cell therapy is substantially compromised due to low cell survival rate and poor local retention post-delivery. These issues drastically limit the application of stem cells for ischemic limb therapy, which requires effective blood perfusion and skeletal muscle regeneration. Herein, based on microfluidic technology, an integrated stem cell and cytokine co-delivery system designed for functional ischemic limb salvage was constructed by first incorporating the myogenic cytokine, fibroblast growth factor 19 (FGF19), into microspheres composed of methacrylate gelatin (GelMA). Then adipose-derived stem cells (ADSCs) were highly absorbed into the porous structure of the microspheres, overcoming the insufficient loading efficiency and activities by conventional encapsulation strategy. The fabricated ADSCs/FGF19@μsphere system demonstrated a uniform size of about 180 μm and a highly porous structure with pore sizes between 20 and 40 μm. The resultant system allowed high doses of ADSCs to be precisely engrafted in the lesion and to survive, and achieved sustained FGF19 release in the ischemic region to facilitate myoblast recruitment and differentiation and myofibrils growth. Furthermore, the combination of ADSCs and FGF19 exhibited a positive synergistic effect which substantially improved the therapeutic benefit of angiogenesis and myogenesis, both in vitro and in vivo. In summary, a stem cell and cytokine co-delivery system with the properties of easy preparation and minimal invasiveness was designed to ensure highly efficient cell delivery, sustained cytokine release, and ultimately realizes effective treatment of ischemic limb regeneration.
Collapse
Affiliation(s)
- Ruihan Wang
- Department of Vascular Surgery, Zhongshan Hospital Fudan University, Shanghai, 200032, PR China
- Institute of Vascular Surgery, Fudan University, Shanghai, 200032, PR China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, PR China
| | - Fangqian Wang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310000, Zhejiang, PR China
| | - Shan Lu
- Department of Vascular Surgery, Zhongshan Hospital Fudan University, Shanghai, 200032, PR China
- Institute of Vascular Surgery, Fudan University, Shanghai, 200032, PR China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, PR China
| | - Bin Gao
- Department of Vascular Surgery, Zhongshan Hospital Fudan University, Shanghai, 200032, PR China
- Institute of Vascular Surgery, Fudan University, Shanghai, 200032, PR China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, PR China
| | - Yuanqing Kan
- Department of Vascular Surgery, Zhongshan Hospital Fudan University, Shanghai, 200032, PR China
- Institute of Vascular Surgery, Fudan University, Shanghai, 200032, PR China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, PR China
| | - Tong Yuan
- Department of Vascular Surgery, Zhongshan Hospital Fudan University, Shanghai, 200032, PR China
- Institute of Vascular Surgery, Fudan University, Shanghai, 200032, PR China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, PR China
| | - Yisheng Xu
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Chen Yuan
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Daqiao Guo
- Department of Vascular Surgery, Zhongshan Hospital Fudan University, Shanghai, 200032, PR China
- Institute of Vascular Surgery, Fudan University, Shanghai, 200032, PR China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, PR China
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital Fudan University, Shanghai, 200032, PR China
- Institute of Vascular Surgery, Fudan University, Shanghai, 200032, PR China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, PR China
| | - Xiaohua Yu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310000, Zhejiang, PR China
| | - Yi Si
- Department of Vascular Surgery, Zhongshan Hospital Fudan University, Shanghai, 200032, PR China
- Institute of Vascular Surgery, Fudan University, Shanghai, 200032, PR China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, PR China
| |
Collapse
|
22
|
Luo S, Yang Z, Chen R, You D, Teng F, Yuan Y, Liu W, Li J, Zhang H. Cytokine receptor-like factor 1 (CRLF1) promotes cardiac fibrosis via ERK1/2 signaling pathway. J Zhejiang Univ Sci B 2023; 24:682-697. [PMID: 37551555 PMCID: PMC10423965 DOI: 10.1631/jzus.b2200506] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/10/2023] [Indexed: 08/09/2023]
Abstract
Cardiac fibrosis is a cause of morbidity and mortality in people with heart disease. Anti-fibrosis treatment is a significant therapy for heart disease, but there is still no thorough understanding of fibrotic mechanisms. This study was carried out to ascertain the functions of cytokine receptor-like factor 1 (CRLF1) in cardiac fibrosis and clarify its regulatory mechanisms. We found that CRLF1 was expressed predominantly in cardiac fibroblasts. Its expression was up-regulated not only in a mouse heart fibrotic model induced by myocardial infarction, but also in mouse and human cardiac fibroblasts provoked by transforming growth factor-β1 (TGF-β1). Gain- and loss-of-function experiments of CRLF1 were carried out in neonatal mice cardiac fibroblasts (NMCFs) with or without TGF-β1 stimulation. CRLF1 overexpression increased cell viability, collagen production, cell proliferation capacity, and myofibroblast transformation of NMCFs with or without TGF-β1 stimulation, while silencing of CRLF1 had the opposite effects. An inhibitor of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway and different inhibitors of TGF-β1 signaling cascades, comprising mothers against decapentaplegic homolog (SMAD)-dependent and SMAD-independent pathways, were applied to investigate the mechanisms involved. CRLF1 exerted its functions by activating the ERK1/2 signaling pathway. Furthermore, the SMAD-dependent pathway, not the SMAD-independent pathway, was responsible for CRLF1 up-regulation in NMCFs treated with TGF-β1. In summary, activation of the TGF-β1/SMAD signaling pathway in cardiac fibrosis increased CRLF1 expression. CRLF1 then aggravated cardiac fibrosis by activating the ERK1/2 signaling pathway. CRLF1 could become a novel potential target for intervention and remedy of cardiac fibrosis.
Collapse
Affiliation(s)
- Shenjian Luo
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhi Yang
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
| | - Ruxin Chen
- State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Danming You
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
| | - Fei Teng
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Youwen Yuan
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenhui Liu
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
| | - Jin Li
- Department of Endocrinology, Shanxi Medical University Affiliated Second Hospital, Taiyuan 030001, China.
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China.
- State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
23
|
Ardizzone A, Bova V, Casili G, Repici A, Lanza M, Giuffrida R, Colarossi C, Mare M, Cuzzocrea S, Esposito E, Paterniti I. Role of Basic Fibroblast Growth Factor in Cancer: Biological Activity, Targeted Therapies, and Prognostic Value. Cells 2023; 12:cells12071002. [PMID: 37048074 PMCID: PMC10093572 DOI: 10.3390/cells12071002] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Cancer is the leading cause of death worldwide; thus, it is necessary to find successful strategies. Several growth factors, such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF, FGF2), and transforming growth factor beta (TGF-β), are involved in the main processes that fuel tumor growth, i.e., cell proliferation, angiogenesis, and metastasis, by activating important signaling pathways, including PLC-γ/PI3/Ca2+ signaling, leading to PKC activation. Here, we focused on bFGF, which, when secreted by tumor cells, mediates several signal transductions and plays an influential role in tumor cells and in the development of chemoresistance. The biological mechanism of bFGF is shown by its interaction with its four receptor subtypes: fibroblast growth factor receptor (FGFR) 1, FGFR2, FGFR3, and FGFR4. The bFGF–FGFR interaction stimulates tumor cell proliferation and invasion, resulting in an upregulation of pro-inflammatory and anti-apoptotic tumor cell proteins. Considering the involvement of the bFGF/FGFR axis in oncogenesis, preclinical and clinical studies have been conducted to develop new therapeutic strategies, alone and/or in combination, aimed at intervening on the bFGF/FGFR axis. Therefore, this review aimed to comprehensively examine the biological mechanisms underlying bFGF in the tumor microenvironment, the different anticancer therapies currently available that target the FGFRs, and the prognostic value of bFGF.
Collapse
Affiliation(s)
- Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| | - Valentina Bova
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| | - Alberto Repici
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| | | | - Cristina Colarossi
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy
| | - Marzia Mare
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
- Correspondence: ; Tel.: +39-090-6765208
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| |
Collapse
|
24
|
Zhu X, Chen H, Li H, Ren H, Ye C, Xu K, Liu J, Du F, Zhang Z, Liu Y, Xie X, Wang M, Ma T, Chong W, Shang L, Li L. ITGB1-mediated molecular landscape and cuproptosis phenotype induced the worse prognosis in diffuse gastric cancer. Front Oncol 2023; 13:1115510. [PMID: 37007126 PMCID: PMC10063208 DOI: 10.3389/fonc.2023.1115510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
Diffuse type gastric cancer was identified with relatively worse prognosis than other Lauren’s histological classification. Integrin β1 (ITGB1) was a member of integrin family which played a markedly important role in tumorigenesis and progression. However, the influence of ITGB1 in diffuse gastric cancer (DGC) remains uncertain. Here, we leveraged the transcriptomic and proteomic data to explore the association between ITGB1 expression and clinicopathologic information and biological process in DGC. Cell phenotype experiments combined with quantitative-PCR (q-PCR) and western blotting were utilized to identify the potential molecular mechanism underling ITGB1.Transcriptomics and proteomics both revealed that the higher ITGB1 expression was significantly associated with worse prognosis in DGC, but not in intestinal GC. Genomic analysis indicated that the mutation frequency of significantly mutated genes of ARID1A and COL11A1, and mutational signatures of SBS6 and SBS15 were markedly increased in the ITGB1 low expression subgroup. The enrichment analysis revealed diverse pathways related to dysregulation of ITGB1 in DGC, especially in cell adhesion, proliferation, metabolism reprogramming, and immune regulation alterations. Elevated activities of kinase-ROCK1, PKACA/PRKACA and AKT1 were observed in the ITGB1 high-expression subgroup. The ssGSEA analysis also found that ITGB1 low-expression had a higher cuproptosis score and was negatively correlated with key regulators of cuproptosis, including FDX1, DLAT, and DLST. We further observed that the upregulated expression of mitochondrial tricarboxylic acid (TCA) cycle in the ITGB1 low-expression group. Reduced expression of ITGB1 inhibited the ability of cell proliferation and motility and also potentiated the cell sensitive to copper ionophores via western blotting assay. Overall, this study revealed that ITGB1 was a protumorigenic gene and regulated tumor metabolism and cuproptosis in DGC.
Collapse
Affiliation(s)
- Xingyu Zhu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hao Chen
- Clinical Research Center of Shandong University, Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Han Li
- Department of Gastroenterological Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Huicheng Ren
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Chunshui Ye
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Kang Xu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jin Liu
- Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Fengying Du
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Zihao Zhang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuan Liu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaozhou Xie
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Mingfei Wang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Tianrong Ma
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Wei Chong
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- *Correspondence: Wei Chong, ; ; Leping Li, ; Liang Shang,
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- *Correspondence: Wei Chong, ; ; Leping Li, ; Liang Shang,
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, China
- Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- *Correspondence: Wei Chong, ; ; Leping Li, ; Liang Shang,
| |
Collapse
|
25
|
Serum Fibroblast Growth Factor 19 as a Biomarker in Hepatitis B Virus-Related Liver Disease. HEPATITIS MONTHLY 2022. [DOI: 10.5812/hepatmon-130652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Background: Past research has found that fibroblast growth factor 19 (FGF19) is associated with several hepatic disorders, such as alcoholic liver disease and primary biliary cirrhosis. However, there is currently a lack of relevant studies on the relationship between FGF19 and hepatitis B virus (HBV)-related liver disease. Objectives: This study aimed to assess the role of serum FGF19 as a new biomarker for HBV-related liver disease and provide scientific data to show the clinical value of this biomarker. Methods: A retrospective study included 37 patients with chronic hepatitis B (CHB), 33 patients with HBV-related cirrhosis (HBV-cirrhosis), and 32 patients with HBV-related hepatocellular carcinoma (HBV-HCC). Furthermore, 33 normal people were randomly selected as healthy controls. The serum levels of FGF19 were measured by ELISA. Results: Serum FGF19 levels were increased sequentially in the CHB group, HBV-cirrhosis group, and HBV-HCC group. Furthermore, serum FGF19 levels positively correlated with alpha-fetoprotein, prothrombin time, international normalized ratio, total bilirubin, direct bilirubin, alanine aminotransferase, aspartate aminotransferase, gamma-glutamyl-transferase, alkaline phosphatase, total bile acid, serum markers for liver fibrosis, ascites, cirrhosis, Child-Pugh classification and model for end-stage liver disease sodium (MELD-Na) score, while negatively correlated with platelet count, prothrombin activity, and albumin. The diagnostic threshold of serum FGF19 for HBV-related HCC was 165.32 pg/mL, with a sensitivity of 81.25% and specificity of 58.57%. Conclusions: Serum FGF19 levels are positively associated with cholestasis, hepatocyte damage, and liver fibrosis but negatively correlated with liver synthetic function and liver functional reserve in HBV-related liver disease. Diverse changes in serum FGF19 may be used as a predictive marker for the progression of HBV-related liver disease. In addition, serum FGF19 has a potential role in monitoring carcinogenesis in patients with HBV-related liver disease.
Collapse
|
26
|
Zhou L, Yi Y, Liu C, Chen Z. Constructing a novel prognostic signature of tumor driver genes for breast cancer. Am J Transl Res 2022; 14:4515-4531. [PMID: 35958490 PMCID: PMC9360863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/27/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVES To systematically explore the function and prognostic ability of tumor-driver genes (TDGs) in breast carcinoma (BRCA). METHODS Functional enrichment analysis of BRCA differentially expressed TDGs was assesed. We used univariate Cox, lasso, and multivariate Cox regression to identify the independent prognostic TDGs of BRCA. Then we constructed a prognostic signature and verified its predictive performance. Gene set enrichment analysis of the signal pathway revealed the differences between the prognostic signature high- and low-risk groups. Finally, a nomogram related to the prognostic model was established and verified. RESULTS A total of 595 differentially expressed TDGs were identified, which are related to various molecular mechanisms of BRCA progression. We identified 8 independent prognostic TDGs for BRCA and validated their expression and prognosis with public data and clinical samples. The BRCA cohort was divided into training and validation cohorts, and prognostic signatures were constructed separately. The log-rank test showed that the survival rate of the high-risk group was significantly lower than that of the low-risk group in the prognostic signature (P<0.001); the AUC in the three cohorts were 0.805, 0.712, and 0.760, respectively; the nomogram also showed better predictive performance. Analyzing the difference between the two risk subtypes, the high-risk group is mainly enriched in angiogenesis, MTORC1, epithelial-mesenchymal transition and glycolysis, which means it is highly malignant. CONCLUSIONS The prognostic signature and nomogram was confirmed to accurately predict the prognosis of patients with BRCA and we validated the hub genes, suggesting their potential as future therapeutic targets.
Collapse
Affiliation(s)
- Liqiang Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Yali Yi
- Department of Oncology, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Chuan Liu
- Key Laboratory of Molecular Medicine of Jiangxi Province, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
| | - Zhiqing Chen
- Key Laboratory of Molecular Medicine of Jiangxi Province, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, China
| |
Collapse
|
27
|
Huang M, Lin Y, Wang C, Deng L, Chen M, Assaraf YG, Chen ZS, Ye W, Zhang D. New insights into antiangiogenic therapy resistance in cancer: Mechanisms and therapeutic aspects. Drug Resist Updat 2022; 64:100849. [PMID: 35842983 DOI: 10.1016/j.drup.2022.100849] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Angiogenesis is a hallmark of cancer and is required for tumor growth and progression. Antiangiogenic therapy has been revolutionarily developing and was approved for the treatment of various types of cancer for nearly two decades, among which bevacizumab and sorafenib continue to be the two most frequently used antiangiogenic drugs. Although antiangiogenic therapy has brought substantial survival benefits to many cancer patients, resistance to antiangiogenic drugs frequently occurs during clinical treatment, leading to poor outcomes and treatment failure. Cumulative evidence has demonstrated that the intricate interplay among tumor cells, bone marrow-derived cells, and local stromal cells critically allows for tumor escape from antiangiogenic therapy. Currently, drug resistance has become the main challenge that hinders the therapeutic efficacies of antiangiogenic therapy. In this review, we describe and summarize the cellular and molecular mechanisms conferring tumor drug resistance to antiangiogenic therapy, which was predominantly associated with redundancy in angiogenic signaling molecules (e.g., VEGFs, GM-CSF, G-CSF, and IL17), alterations in biological processes of tumor cells (e.g., tumor invasiveness and metastasis, stemness, autophagy, metabolic reprogramming, vessel co-option, and vasculogenic mimicry), increased recruitment of bone marrow-derived cells (e.g., myeloid-derived suppressive cells, tumor-associated macrophages, and tumor-associated neutrophils), and changes in the biological functions and features of local stromal cells (e.g., pericytes, cancer-associated fibroblasts, and endothelial cells). We also review potential biomarkers to predict the response to antiangiogenic therapy in cancer patients, which mainly consist of imaging biomarkers, cellular and extracellular proteins, a certain type of bone marrow-derived cells, local stromal cell content (e.g., pericyte coverage) as well as serum or plasma biomarkers (e.g., non-coding RNAs). Finally, we highlight the recent advances in combination strategies with the aim of enhancing the response to antiangiogenic therapy in cancer patients and mouse models. This review introduces a comprehensive understanding of the mechanisms and biomarkers associated with the evasion of antiangiogenic therapy in cancer, providing an outlook for developing more effective approaches to promote the therapeutic efficacy of antiangiogenic therapy.
Collapse
Affiliation(s)
- Maohua Huang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Yuning Lin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Chenran Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Lijuan Deng
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Minfeng Chen
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Institute for Biotechnology, St. John's University, NY 11439, USA.
| | - Wencai Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Dongmei Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
28
|
Hegde M, Daimary UD, Jose S, Sajeev A, Chinnathambi A, Alharbi SA, Shakibaei M, Kunnumakkara AB. Differential Expression of Genes Regulating Store-operated Calcium Entry in Conjunction With Mitochondrial Dynamics as Potential Biomarkers for Cancer: A Single-Cell RNA Analysis. Front Genet 2022; 13:866473. [PMID: 35711942 PMCID: PMC9197647 DOI: 10.3389/fgene.2022.866473] [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: 01/31/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Regulation of intracellular concentration of calcium levels is crucial for cell signaling, homeostasis, and in the pathology of diseases including cancer. Agonist-induced entry of calcium ions into the non-excitable cells is mediated by store-operated calcium channels (SOCs). This pathway is activated by the release of calcium ions from the endoplasmic reticulum and further regulated by the calcium uptake through mitochondria leading to calcium-dependent inactivation of calcium-release activated calcium channels (CARC). SOCs including stromal interaction molecules (STIM) and ORAI proteins have been implicated in tumor growth, progression, and metastasis. In the present study, we analyzed the mRNA and protein expression of genes mediating SOCs-STIM1, STIM2, ORAI1, ORAI2, ORAI3, TRPC1, TRPC3, TRPC4, TRPC5, TRPC6, TRPC7, TRPV1, TRPV2, TRPM1, and TRPM7 in head and neck squamous cell cancer (HNSC) patients using TCGA and CPTAC analysis. Further, our in silico analysis showed a significant correlation between the expression of SOCs and genes involved in the mitochondrial dynamics (MDGs) both at mRNA and protein levels. Protein-protein docking results showed lower binding energy for SOCs with MDGs. Subsequently, we validated these results using gene expression and single-cell RNA sequencing datasets retrieved from Gene Expression Omnibus (GEO). Single-cell gene expression analysis of HNSC tumor tissues revealed that SOCs expression is remarkably associated with the MDGs expression in both cancer and fibroblast cells.
Collapse
Affiliation(s)
- Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati, India.,DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati, India
| | - Uzini Devi Daimary
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati, India.,DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati, India
| | - Sandra Jose
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati, India.,DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati, India
| | - Anjana Sajeev
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati, India.,DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati, India
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati, India.,DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati, India
| |
Collapse
|
29
|
Store-Operated Calcium Entry and Its Implications in Cancer Stem Cells. Cells 2022; 11:cells11081332. [PMID: 35456011 PMCID: PMC9032688 DOI: 10.3390/cells11081332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/12/2022] [Indexed: 12/25/2022] Open
Abstract
Tumors are composed by a heterogeneous population of cells. Among them, a sub-population of cells, termed cancer stem cells, exhibit stemness features, such as self-renewal capabilities, disposition to differentiate to a more proliferative state, and chemotherapy resistance, processes that are all mediated by Ca2+. Ca2+ homeostasis is vital for several physiological processes, and alterations in the patterns of expressions of the proteins and molecules that modulate it have recently become a cancer hallmark. Store-operated Ca2+ entry is a major mechanism for Ca2+ entry from the extracellular medium in non-excitable cells that leads to increases in the cytosolic Ca2+ concentration required for several processes, including cancer stem cell properties. Here, we focus on the participation of STIM, Orai, and TRPC proteins, the store-operated Ca2+ entry key components, in cancer stem cell biology and tumorigenesis.
Collapse
|
30
|
Li TT, Mou J, Pan YJ, Huo FC, Du WQ, Liang J, Wang Y, Zhang LS, Pei DS. MicroRNA-138-1-3p sensitizes sorafenib to hepatocellular carcinoma by targeting PAK5 mediated β-catenin/ABCB1 signaling pathway. J Biomed Sci 2021; 28:56. [PMID: 34340705 PMCID: PMC8327419 DOI: 10.1186/s12929-021-00752-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 07/20/2021] [Indexed: 12/19/2022] Open
Abstract
Background Sorafenib is a kinase inhibitor that is used as a first-line therapy in advanced hepatocellular carcinoma (HCC) patients. However, the existence of sorafenib resistance has limited its therapeutic effect. Through RNA sequencing, we demonstrated that miR-138-1-3p was downregulated in sorafenib resistant HCC cell lines. This study aimed to investigate the role of miR-138-1-3p in sorafenib resistance of HCC. Methods In this study, quantitative real-time PCR (qPCR) and Western Blot were utilized to detect the levels of PAK5 in sorafenib-resistant HCC cells and parental cells. The biological functions of miR-138-1-3p and PAK5 in sorafenib-resistant cells and their parental cells were explored by cell viability assays and flow cytometric analyses. The mechanisms for the involvement of PAK5 were examined via co-immunoprecipitation (co-IP), immunofluorescence, dual luciferase reporter assay and chromatin immunoprecipitation (ChIP). The effects of miR-138-1-3p and PAK5 on HCC sorafenib resistant characteristics were investigated by a xenotransplantation model. Results We detected significant down-regulation of miR-138-1-3p and up-regulation of PAK5 in sorafenib-resistance HCC cell lines. Mechanistic studies revealed that miR-138-1-3p reduced the protein expression of PAK5 by directly targeting the 3′-UTR of PAK5 mRNA. In addition, we verified that PAK5 enhanced the phosphorylation and nuclear translocation of β-catenin that increased the transcriptional activity of a multidrug resistance protein ABCB1. Conclusions PAK5 contributed to the sorafenib resistant characteristics of HCC via β-catenin/ABCB1 signaling pathway. Our findings identified the correlation between miR-138-1-3p and PAK5 and the molecular mechanisms of PAK5-mediated sorafenib resistance in HCC, which provided a potential therapeutic target in advanced HCC patients.
Collapse
Affiliation(s)
- Tong-Tong Li
- Department of Pathology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, People's Republic of China.,Department of Pathology and Pathophysiology, Jiangsu Vocational College of Medicine, Yancheng, 224005, Jiangsu, China
| | - Jie Mou
- Jiangsu Key Laboratory of New Drug and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221006, China
| | - Yao-Jie Pan
- Department of Pathology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Fu-Chun Huo
- Department of Pathology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Wen-Qi Du
- Department of Pathology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Jia Liang
- Department of Pathology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Yang Wang
- Department of Pathology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Lan-Sheng Zhang
- Department of Oncological Radiotherapy, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Dong-Sheng Pei
- Department of Pathology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, People's Republic of China.
| |
Collapse
|
31
|
Terrié E, Déliot N, Benzidane Y, Harnois T, Cousin L, Bois P, Oliver L, Arnault P, Vallette F, Constantin B, Coronas V. Store-Operated Calcium Channels Control Proliferation and Self-Renewal of Cancer Stem Cells from Glioblastoma. Cancers (Basel) 2021; 13:cancers13143428. [PMID: 34298643 PMCID: PMC8307764 DOI: 10.3390/cancers13143428] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/05/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Glioblastoma is a high-grade primary brain tumor that contains a subpopulation of cells called glioblastoma stem cells, which are responsible for tumor initiation, growth and recurrence after treatment. Recent transcriptomic studies have highlighted that calcium pathways predominate in glioblastoma stem cells. Calcium channels have the ability to transduce signals from the microenvironment and are therefore ideally placed to control cellular behavior. Using multiple approaches, we demonstrate in five different primary cultures, previously derived from surgical specimens, that glioblastoma stem cells express store-operated channels (SOC) that support calcium entry into these cells. Pharmacological inhibition of SOC dramatically reduces cell proliferation and stem cell self-renewal in these cultures. By identifying SOC as a critical mechanism involved in the maintenance of the stem cell population in glioblastoma, our study will contribute to the framework for the identification of new therapies against this deadly tumor. Abstract Glioblastoma is the most frequent and deadly form of primary brain tumors. Despite multimodal treatment, more than 90% of patients experience tumor recurrence. Glioblastoma contains a small population of cells, called glioblastoma stem cells (GSC) that are highly resistant to treatment and endowed with the ability to regenerate the tumor, which accounts for tumor recurrence. Transcriptomic studies disclosed an enrichment of calcium (Ca2+) signaling transcripts in GSC. In non-excitable cells, store-operated channels (SOC) represent a major route of Ca2+ influx. As SOC regulate the self-renewal of adult neural stem cells that are possible cells of origin of GSC, we analyzed the roles of SOC in cultures of GSC previously derived from five different glioblastoma surgical specimens. Immunoblotting and immunocytochemistry experiments showed that GSC express Orai1 and TRPC1, two core SOC proteins, along with their activator STIM1. Ca2+ imaging demonstrated that SOC support Ca2+ entries in GSC. Pharmacological inhibition of SOC-dependent Ca2+ entries decreased proliferation, impaired self-renewal, and reduced expression of the stem cell marker SOX2 in GSC. Our data showing the ability of SOC inhibitors to impede GSC self-renewal paves the way for a strategy to target the cells considered responsible for conveying resistance to treatment and tumor relapse.
Collapse
Affiliation(s)
- Elodie Terrié
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
| | - Nadine Déliot
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
| | - Yassine Benzidane
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
| | - Thomas Harnois
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
| | - Laëtitia Cousin
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
| | - Patrick Bois
- EA 4379, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France;
| | - Lisa Oliver
- CRCINA-UMR 1232 INSERM, Université de Nantes, CEDEX 01, 44007 Nantes, France; (L.O.); (F.V.)
| | - Patricia Arnault
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
| | - François Vallette
- CRCINA-UMR 1232 INSERM, Université de Nantes, CEDEX 01, 44007 Nantes, France; (L.O.); (F.V.)
- CNRS GDR3697, Micronit “Microenvironment of Tumor Niches”, 37000 Tours, France
| | - Bruno Constantin
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
- CNRS GDR3697, Micronit “Microenvironment of Tumor Niches”, 37000 Tours, France
| | - Valérie Coronas
- CNRS ERL 7003, Signalisation et Transports Ioniques Membranaires, University of Poitiers, CEDEX 09, 86073 Poitiers, France; (E.T.); (N.D.); (Y.B.); (T.H.); (L.C.); (P.A.); (B.C.)
- CNRS GDR3697, Micronit “Microenvironment of Tumor Niches”, 37000 Tours, France
- Correspondence: ; Tel.: +33-(0)5-49-45-36-55
| |
Collapse
|