1
|
Zhang F, Zhou C, Wang X, Liu Y, Hou Y, Niu L. INHBA, transcriptionally activated by SPI1, facilitates gastric cancer progression by inducing macrophage recruitment and M2 polarization via activating the TGF-β signaling to increase CCL2. Pathol Res Pract 2025; 269:155920. [PMID: 40132395 DOI: 10.1016/j.prp.2025.155920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
Tumor-associated macrophages (TAMs) are associated with the occurrence, development, and poor prognosis of human cancers. Inhibin beta A subunit (INHBA) is found to be aberrantly upregulated in gastric cancer (GC). However, whether INHBA is involved in macrophage recruitment and M2 polarization is unclear. Herein, INHBA expression was increased in GC tumor tissues and cells. INHBA expression was positively correlated with macrophage infiltration and M2 macrophage markers. Knockdown of INHBA in GC cells suppressed macrophage recruitment and M2 polarization by downregulaitng CCL2 expression and secretion. Mechanistic assays showed that SPI1 could bind to INHBA and transcriptionally activate its expression. SPI1 promoted macrophage recruitment and M2 polarization by upregulating INHBA expression. Moreover, SPI1 induced CCL2 expression by regulating INHBA in GC cells. INHBA upregulated CCL2 expression by activating the TGF-β signaling. Furthermore, SPI1-induced macrophages facilitated cell proliferation, migration, and invasion by increasing INHBA expression. INHBA-induced macrophages promoted cell proliferation, migration, and invasion by inducing CCL2 expression. Additionally, knockdown of INHBA inhibited tumor growth in vivo. In conclusion, SPI1 induces the macrophage recruitment and M2 polarization by transcriptionally regulating INHBA to activating the TGF-β signaling, thereby upregulating CCL2 expression and then contributing to GC cell malignant progression. Targeting SPI1/INHBA/CCL2 axis might be a promising therapeutic strategy for GC and potentially used for cancer immunotherapy.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Congya Zhou
- Department of Radiotherapy, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Xifang Wang
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Ying Liu
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Yinyin Hou
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Lu Niu
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an 710068, China.
| |
Collapse
|
2
|
Shen Z, Li M, Zhu H, Song T. TBP activates DCBLD1 transcription to promote cell cycle progression in cervical cancer. Funct Integr Genomics 2024; 24:221. [PMID: 39586885 DOI: 10.1007/s10142-024-01496-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/26/2024] [Accepted: 11/03/2024] [Indexed: 11/27/2024]
Abstract
Discoidin, CUB, and LCCL domain-containing (DCBLD) proteins have been associated with poor prognosis of human cancers. This study investigated the function of DCBLD1 in the development of cervical cancer (CC) and explored its associated mechanism. DCBLD1 was identified as a dysregulated gene in CC via bioinformatics analysis. Immunohistochemistry and RT-qPCR assays revealed increased DCBLD1 expression in CC specimens and cells. Artificial DCBLD1 knockdown blocked the proliferation, invasion, and migration of cells, while promoting cell apoptosis and inducing cell cycle arrest in the G1 phase. Following bioinformatic predictions and subsequent chromatin-immunoprecipitation and luciferase reporter assays, TATA-box binding protein (TBP) was found to be a transcription factor that binds to the DCBLD1 promoter region for transcriptional activation. Knockdown of TBP similarly blocked the malignant properties of CC cells and induced cell cycle arrest, but these changes were reversed by further DCBLD1 overexpression. Xenograft mouse tumors were generated for in vivo validation. Consistently, the tumorigenic activity of CC cells in nude mice was suppressed by TBP knockdown, but restored by DCBLD1 overexpression. In conclusion, this study provides novel evidence that TBP-mediated DCBLD1 activation is correlated with cell cycle and CC progression. TBP and DCBLD1 may serve as potential therapeutic targets for CC management.
Collapse
Affiliation(s)
- Zhigang Shen
- Department of Pharmacy, Jilin Cancer Hospital, NO. 1066, Jinhu Road, High-tech District, Changchun, Jilin, 130012, P.R. China
| | - Mei Li
- Department of Pharmacy, Jilin Cancer Hospital, NO. 1066, Jinhu Road, High-tech District, Changchun, Jilin, 130012, P.R. China
| | - He Zhu
- Department of Gynecology and Oncology, The Second Affiliated Hospital of Jilin University, Changchun, Jilin, 130041, P.R. China
| | - Tao Song
- Department of Pharmacy, Jilin Cancer Hospital, NO. 1066, Jinhu Road, High-tech District, Changchun, Jilin, 130012, P.R. China.
| |
Collapse
|
3
|
Dakal TC, George N, Xu C, Suravajhala P, Kumar A. Predictive and Prognostic Relevance of Tumor-Infiltrating Immune Cells: Tailoring Personalized Treatments against Different Cancer Types. Cancers (Basel) 2024; 16:1626. [PMID: 38730579 PMCID: PMC11082991 DOI: 10.3390/cancers16091626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024] Open
Abstract
TIICs are critical components of the TME and are used to estimate prognostic and treatment responses in many malignancies. TIICs in the tumor microenvironment are assessed and quantified by categorizing immune cells into three subtypes: CD66b+ tumor-associated neutrophils (TANs), FoxP3+ regulatory T cells (Tregs), and CD163+ tumor-associated macrophages (TAMs). In addition, many cancers have tumor-infiltrating M1 and M2 macrophages, neutrophils (Neu), CD4+ T cells (T-helper), CD8+ T cells (T-cytotoxic), eosinophils, and mast cells. A variety of clinical treatments have linked tumor immune cell infiltration (ICI) to immunotherapy receptivity and prognosis. To improve the therapeutic effectiveness of immune-modulating drugs in a wider cancer patient population, immune cells and their interactions in the TME must be better understood. This study examines the clinicopathological effects of TIICs in overcoming tumor-mediated immunosuppression to boost antitumor immune responses and improve cancer prognosis. We successfully analyzed the predictive and prognostic usefulness of TIICs alongside TMB and ICI scores to identify cancer's varied immune landscapes. Traditionally, immune cell infiltration was quantified using flow cytometry, immunohistochemistry, gene set enrichment analysis (GSEA), CIBERSORT, ESTIMATE, and other platforms that use integrated immune gene sets from previously published studies. We have also thoroughly examined traditional limitations and newly created unsupervised clustering and deconvolution techniques (SpatialVizScore and ProTICS). These methods predict patient outcomes and treatment responses better. These models may also identify individuals who may benefit more from adjuvant or neoadjuvant treatment. Overall, we think that the significant contribution of TIICs in cancer will greatly benefit postoperative follow-up, therapy, interventions, and informed choices on customized cancer medicines.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur 313001, Rajasthan, India
| | - Nancy George
- Department of Biotechnology, Chandigarh University, Mohali 140413, Punjab, India;
| | - Caiming Xu
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of the City of Hope, Monrovia, CA 91010, USA;
| | - Prashanth Suravajhala
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana P.O. 690525, Kerala, India;
| | - Abhishek Kumar
- Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, Karnataka, India
| |
Collapse
|
4
|
Wu Z, Chen J, Yang L, Sun K, Jiang Q, Dong F, Lu W, Chen R, Chen Y. Elevated INHBA Promotes Tumor Progression of Cervical Cancer. Technol Cancer Res Treat 2024; 23:15330338241234798. [PMID: 38419562 PMCID: PMC10903216 DOI: 10.1177/15330338241234798] [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] [Indexed: 03/02/2024] Open
Abstract
Objectives: This study aimed to explore the role of inhibin subunit beta A (INHBA) in the progression of cervical cancer (CCa) and investigate its potential as a therapeutic target. Specifically, the objectives were to assess the expression levels of INHBA in CCa, examine its correlation with patient survival, and elucidate its impact on CCa cell proliferation, cell cycle regulation, migration, invasion, and in vivo tumor growth and metastasis. Methods: To achieve the objectives, we conducted a comprehensive set of experimental methods. INHBA expression in CCa was analyzed, and its association with patient survival was assessed using clinical data. In vitro experiments involved the investigation of INHBA's effects on CCa cell proliferation, cell cycle dynamics, migration, and invasion through the epithelial-mesenchymal transition (EMT) process. Additionally, in vivo experiments were performed to evaluate the influence of INHBA on CCa growth and lung metastasis. Results: The results of this study revealed upregulated expression of INHBA in CCa, with a significant association between high INHBA expression and poor patient survival. Functionally, INHBA was found to promote the proliferation of CCa cells, regulate the cell cycle, and enhance migration and invasion through the EMT process in vitro. Moreover, in vivo experiments demonstrated that INHBA facilitated the growth and lung metastasis of CCa. Conclusion: In conclusion, our findings suggest that INHBA plays a crucial role in the progression of cervical cancer. The upregulation of INHBA is associated with poor patient survival, and its involvement in promoting key aspects of cancer progression makes it a potential therapeutic target for CCa treatment. These results provide valuable insights into the molecular mechanisms underlying CCa and offer a foundation for further exploration of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Zhenghong Wu
- Department of Gynecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| | - Jina Chen
- Department of Gynecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| | - Lina Yang
- Department of Gynecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| | - Ke Sun
- Department of Gynecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| | - Qianqian Jiang
- Department of Gynecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| | - Fuyun Dong
- Department of Gynecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| | - Wei Lu
- Department of Gynecology, Shanghai Electric Power Hospital, Shanghai, P.R. China
| | - Rujun Chen
- Department of Gynecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| | - Yaping Chen
- Department of Gynecology and Obstetrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|