1
|
Qu H, Zhao J, Zuo X, He H, Wang X, Li H, Zhang K. TGF-β-mediated activation of fibroblasts in cervical cancer: implications for tumor microenvironment and prognosis. PeerJ 2025; 13:e19072. [PMID: 40124621 PMCID: PMC11929507 DOI: 10.7717/peerj.19072] [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: 10/29/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Background Cervical cancer (CC) is a prevalent female malignancy strongly influenced by the tumor microenvironment (TME). This study focuses on the role of TGF-β signaling in cancer-associated fibroblasts (CAFs) and its interaction with immune cells, aiming to elucidate its impact on CC progression. Methods The TME of CC patients was analyzed using scRNA-seq data and we identified the major cell types in the TME with a focus on the activation of the TGF-β signaling pathway in fibroblasts. Gene modules related to the TGF-β signaling pathway were identified by Weighted correlation network analysis (WGCNA). Using The Cancer Genome Atlas Cervical Squamous Cell Carcinoma and Endocervical Adenocarcinoma (TCGA-CESC) dataset, a prognostic gene model was constructed by univariate Cox, LASSO Cox and multivariate Cox regression analyses. For cellular validation, the mRNA level of prognostic model-related genes was tested via quantitative real-time real-time polymerase chain reaction (PCR). Thereafter, the following assays, including cell counting kit-8, scratch and wound healing assays, were applied to assess the viability, migration and invasion of CC cells. Results Analysis at single-cell resolution identified nine major cell types in the TME, and significant activation of the TGF-β signaling pathway in fibroblasts was correlated with tumor proliferation and differentiation. Strong TGF-β signaling communication between fibroblasts and macrophages and NK/T cells suggested a crucial role in the shaping of the immunosuppressive microenvironment. WGCNA analysis identified gene modules significantly associated with the TGF-β signaling pathway. The prognostic model constructed based on three genes, ITGA5, SHF and SNRPN, demonstrated good predictive ability in multiple datasets, validating its potential for clinical application. Meanwhile, the cellular validation assays have revealed the higher expression of ITGA5 and SNRPN and lower expression of SHF in CC cells. Further, ITGA5 knockdown suppressed the viability, migration and invasion of CC cells. Conclusion This study confirmed the important role of the TGF-β signaling pathway in CC, especially in fibroblasts on tumor microenvironment and tumor progression. The current model could effectively evaluate the prognosis of CC, providing a theoretical foundation for developing CC therapies according to the TGF-β signaling pathway. The present results provide new perspectives for further research on the pathological mechanisms and clinical management of CC.
Collapse
Affiliation(s)
- Haina Qu
- Obstetrics and Gynecology Department, Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Jing Zhao
- Obstetrics and Gynecology Department, Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Xia Zuo
- Obstetrics and Gynecology Department, Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Hongyue He
- Obstetrics and Gynecology Department, Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Xiaohan Wang
- Obstetrics and Gynecology Department, Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Huiyan Li
- Obstetrics and Gynecology Department, Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Kun Zhang
- Obstetrics and Gynecology Department, Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| |
Collapse
|
2
|
Trugilo KP, Cebinelli GCM, Castilha EP, da Silva MR, Berti FCB, de Oliveira KB. The role of transforming growth factor β in cervical carcinogenesis. Cytokine Growth Factor Rev 2024; 80:12-23. [PMID: 39482191 DOI: 10.1016/j.cytogfr.2024.10.006] [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: 08/23/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024]
Abstract
Human papillomavirus (HPV) is involved in virtually all cases of cervical cancer. However, HPV alone is not sufficient to cause malignant development. The effects of chronic inflammation and the interaction of immune components with the microenvironment infected with the high-risk HPV type (HR) may contribute to cancer development. Transforming growth factor β (TGFB) appears to play an important role in cervical carcinogenesis. Protein and mRNA levels of this cytokine gradually increase as normal tissue develops into malignant tissue and are closely related to the severity of HPV infection. At the onset of infection, TGFB can inhibit the proliferation of infected cells and viral amplification by inhibiting cell growth and downregulating the transcriptional activity of the long control region (LCR) of HPV, thereby reducing the expression of early genes. When infected cells progress to a malignant phenotype, the response to the cell growth inhibitory effect of TGFB1 is lost and the suppression of E6 and E7 expression decreases. Subsequently, TGFB1 expression is upregulated by high levels of E6 and E7 oncoproteins, leading to an increase in TGFB1 in the tumor microenvironment, where this molecule promotes epithelial-to-mesenchymal transition (EMT), cell motility, angiogenesis, and immunosuppression. This interaction between HPV oncoproteins and TGFB1 is an important mechanism promoting the development and progression of cervical cancer.
Collapse
Affiliation(s)
- Kleber Paiva Trugilo
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, PR 86057-970, Brazil.
| | | | - Eliza Pizarro Castilha
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, PR 86057-970, Brazil.
| | - Mariane Ricciardi da Silva
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, PR 86057-970, Brazil.
| | | | - Karen Brajão de Oliveira
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, PR 86057-970, Brazil.
| |
Collapse
|
3
|
Kromidas E, Geier A, Weghofer A, Liu HY, Weiss M, Loskill P. Immunocompetent PDMS-Free Organ-on-Chip Model of Cervical Cancer Integrating Patient-Specific Cervical Fibroblasts and Neutrophils. Adv Healthc Mater 2024; 13:e2302714. [PMID: 38029413 DOI: 10.1002/adhm.202302714] [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: 08/17/2023] [Revised: 11/04/2023] [Indexed: 12/01/2023]
Abstract
Despite preventive measures and available treatments, cervical cancer still ranks as the fourth most prevalent cancer among women worldwide and remains the leading cause of cancer death in women in many developing countries. To gain further insights into pathogenesis and to develop novel (immuno)therapies, more sophisticated human models recreating patient heterogeneities and including aspects of the tumor microenvironment are urgently required. A novel polydimethylsiloxane-free microfluidic platform, designed specifically for the generation and ccultivation of cervical cancerous tissue, is introduced. The microscale open-top tissue chambers of the cervical cancer-on-chip (CCoC) enable facile generation and long-term cultivation of SiHa spheroids in co-culture with donor-derived cervical fibroblasts. The resulting 3D tissue emulates physiological architecture and allows dissection of distinct effects of the stromal tissue on cancer viability and growth. Treatment with cisplatin at clinically-relevant routes of administration and dosing highlights the platform's applicability for drug testing. Moreover, the model is amenable for integration and recruitment of donor-derived neutrophils from the microvasculature-like channel into the tissue, all while retaining their ability to produce neutrophil extracellular traps. In the future, the immunocompetent CCoC featuring donor-specific primary cells and tumor spheroids has the potential to contribute to the development of new (immuno)therapeutic options.
Collapse
Affiliation(s)
- Elena Kromidas
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, 72074, Tübingen, Germany
| | - Alicia Geier
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, 72074, Tübingen, Germany
| | - Adrian Weghofer
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, 72074, Tübingen, Germany
| | - Hui-Yu Liu
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, 72074, Tübingen, Germany
| | - Martin Weiss
- Department for Biomedicine and Materials Science, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
- Department for Women's Health, Faculty of Medicine, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Peter Loskill
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, 72074, Tübingen, Germany
- Department for Biomedicine and Materials Science, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
- 3R Center Tübingen for In Vitro Models and Alternatives to Animal Testing, 72074, Tübingen, Germany
| |
Collapse
|
4
|
Bueno-Urquiza LJ, Godínez-Rubí M, Villegas-Pineda JC, Vega-Magaña AN, Jave-Suárez LF, Puebla-Mora AG, Aguirre-Sandoval GE, Martínez-Silva MG, Ramírez-de-Arellano A, Pereira-Suárez AL. Phenotypic Heterogeneity of Cancer Associated Fibroblasts in Cervical Cancer Progression: FAP as a Central Activation Marker. Cells 2024; 13:560. [PMID: 38606999 PMCID: PMC11010959 DOI: 10.3390/cells13070560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/09/2024] [Accepted: 03/10/2024] [Indexed: 04/13/2024] Open
Abstract
Cervical cancer (CC) is the fourth leading cancer among women and is one of the principal gynecological malignancies. In the tumor microenvironment, cancer-associated fibroblasts (CAFs) play a crucial role during malignant progression, exhibiting a variety of heterogeneous phenotypes. CAFs express phenotypic markers like fibroblast activation protein (FAP), vimentin, S100A4, α-smooth muscle actin (αSMA), and functional markers such as MMP9. This study aimed to evaluate the protein expression of vimentin, S100A4, αSMA, FAP, and MMP9 in mesenchymal stem cells (MSC)-CAF cells, as well as in cervical cancer samples. MSC cells were stimulated with HeLa and SiHa tumor cell supernatants, followed by protein evaluation and cytokine profile to confirm differentiation towards a CAF phenotype. In addition, automated immunohistochemistry (IHQa) was performed to evaluate the expression of these proteins in CC samples at different stages. Our findings revealed a high expression of FAP in stimulated MSC cells, accompanied by the secretion of pro/anti-inflammatory cytokines. In the other hand, CC samples were observed to have high expression of FAP, vimentin, αSMA, and MMP9. Most importantly, there was a high expression of their activation proteins αSMA and FAP during the different stages. In the early stages, a myofibroblast-like phenotype (CAFs αSMA+ FAP+), and in the late stages a protumoral phenotype (CAF αSMA- FAP+). In summary, FAP has a crucial role in the activation of CAFs during cervical cancer progression.
Collapse
Affiliation(s)
- Lesly Jazmin Bueno-Urquiza
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (L.J.B.-U.); (A.N.V.-M.); (A.R.-d.-A.)
| | - Marisol Godínez-Rubí
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (M.G.-R.); (J.C.V.-P.); (A.G.P.-M.); (G.E.A.-S.)
| | - Julio César Villegas-Pineda
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (M.G.-R.); (J.C.V.-P.); (A.G.P.-M.); (G.E.A.-S.)
| | - Alejandra Natali Vega-Magaña
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (L.J.B.-U.); (A.N.V.-M.); (A.R.-d.-A.)
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (M.G.-R.); (J.C.V.-P.); (A.G.P.-M.); (G.E.A.-S.)
| | - Luis Felipe Jave-Suárez
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico;
| | - Ana Graciela Puebla-Mora
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (M.G.-R.); (J.C.V.-P.); (A.G.P.-M.); (G.E.A.-S.)
| | - Gloria Estefanía Aguirre-Sandoval
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (M.G.-R.); (J.C.V.-P.); (A.G.P.-M.); (G.E.A.-S.)
| | - María Guadalupe Martínez-Silva
- Departamento de Anatomía Patológica, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social (IMSS), Guadalajara 44340, Mexico;
| | - Adrián Ramírez-de-Arellano
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (L.J.B.-U.); (A.N.V.-M.); (A.R.-d.-A.)
| | - Ana Laura Pereira-Suárez
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (L.J.B.-U.); (A.N.V.-M.); (A.R.-d.-A.)
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (M.G.-R.); (J.C.V.-P.); (A.G.P.-M.); (G.E.A.-S.)
| |
Collapse
|
5
|
Duggins-Warf M, Ghalali A, Sesen J, Martinez T, Fehnel KP, Pineda S, Zurakowski D, Smith ER. Disease specific urinary biomarkers in the central nervous system. Sci Rep 2023; 13:19244. [PMID: 37935834 PMCID: PMC10630515 DOI: 10.1038/s41598-023-46763-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 11/04/2023] [Indexed: 11/09/2023] Open
Abstract
Urinary biomarkers can diagnose and monitor pathophysiologic conditions in the central nervous system (CNS). However, focus is often on single diseases, with limited data on discriminatory capability of this approach in a general setting. Here, we demonstrate that different classes of CNS disease exhibit distinct biomarker patterns, evidence of disease-specific "fingerprinting." Urine from 218 patients with pathology-confirmed tumors or cerebrovascular disease, controls (n = 33) were collected. ELISA and/or bead-based multiplexing quantified levels of 21 putative urinary biomarkers. Analysis identified biomarkers capable of distinguishing each disease from controls and other diseases. Mann-Whitney U tests identified biomarkers with differential expression between disease types and controls (P ≤ 0.001). Subsequent receiver-operating characteristic (ROC) analyses revealed distinguishing biomarkers with high sensitivity and specificity. Areas under the curve (AUCs) ranged 0.8563-1.000 (P values ≤ 0.0003), sensitivities ranged 80.00-100.00%, and specificities ranged 80.95-100.00%. These data demonstrate proof-of-principle evidence that disease-specific urinary biomarker signatures exist. In contrast to non-specific responses to ischemia or injury, these results suggest that urinary biomarkers accurately reflect unique biological processes distinct to different diseases. This work can be used to generate disease-specific panels for enhancing diagnosis, assisting less-invasive follow-up and herald utility by revealing putative disease-specific therapeutic targets.
Collapse
Affiliation(s)
- Micah Duggins-Warf
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA
| | - Aram Ghalali
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA
| | - Julie Sesen
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA
| | - Tyra Martinez
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA
| | - Katie P Fehnel
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA
| | - Steven Pineda
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA
| | - David Zurakowski
- Department of Surgery, Boston Children's Hospital, Boston, MA, USA
| | - Edward R Smith
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA.
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
6
|
Liu C, Zhang M, Yan X, Ni Y, Gong Y, Wang C, Zhang X, Wan L, Yang H, Ge C, Li Y, Zou W, Huang R, Li X, Sun B, Liu B, Yue J, Yu J. Single-cell dissection of cellular and molecular features underlying human cervical squamous cell carcinoma initiation and progression. SCIENCE ADVANCES 2023; 9:eadd8977. [PMID: 36706185 PMCID: PMC9882988 DOI: 10.1126/sciadv.add8977] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 12/23/2022] [Indexed: 05/28/2023]
Abstract
Cervical squamous cell carcinoma (CESC) is a prototypical human cancer with well-characterized pathological stages of initiation and progression. However, high-resolution knowledge of the transcriptional programs underlying each stage of CESC is lacking, and important questions remain. We performed single-cell RNA sequencing of 76,911 individual cells from 13 samples of human cervical tissues at various stages of malignancy, illuminating the transcriptional tumorigenic trajectory of cervical epithelial cells and revealing key factors involved in CESC initiation and progression. In addition, we found significant correlations between the abundance of specific myeloid, lymphoid, and endothelial cell populations and the progression of CESC, which were also associated with patients' prognosis. Last, we demonstrated the tumor-promoting function of matrix cancer-associated fibroblasts via the NRG1-ERBB3 pathway in CESC. This study provides a valuable resource and deeper insights into CESC initiation and progression, which is helpful in refining CESC diagnosis and for the design of optimal treatment strategies.
Collapse
Affiliation(s)
- Chao Liu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan 250117, China
| | - Min Zhang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Xinlong Yan
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing 100124, China
| | - Yanli Ni
- State Key Laboratory of Experimental Hematology, Senior Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Yandong Gong
- State Key Laboratory of Experimental Hematology, Senior Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Cong Wang
- Department of Gynecologic Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiaoling Zhang
- Department of Gynecologic Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Lingfei Wan
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing 100124, China
| | - Hui Yang
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing 100124, China
| | - Chen Ge
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing 100124, China
| | - Yunqiao Li
- State Key Laboratory of Experimental Hematology, Senior Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Wenxue Zou
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Rui Huang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiaohui Li
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Bing Sun
- Department of Radiation Oncology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Bing Liu
- State Key Laboratory of Experimental Hematology, Senior Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Jinbo Yue
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Jinming Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan 250117, China
| |
Collapse
|
7
|
Villegas-Pineda JC, Ramírez-de-Arellano A, Bueno-Urquiza LJ, Lizarazo-Taborda MDR, Pereira-Suárez AL. Cancer-associated fibroblasts in gynecological malignancies: are they really allies of the enemy? Front Oncol 2023; 13:1106757. [PMID: 37168385 PMCID: PMC10164963 DOI: 10.3389/fonc.2023.1106757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/24/2023] [Indexed: 05/13/2023] Open
Abstract
Molecular and cellular components of the tumor microenvironment are essential for cancer progression. The cellular element comprises cancer cells and heterogeneous populations of non-cancer cells that satisfy tumor needs. Immune, vascular, and mesenchymal cells provide the necessary factors to feed the tumor mass, promote its development, and favor the spread of cancer cells from the primary site to adjacent and distant anatomical sites. Cancer-associated fibroblasts (CAFs) are mesenchymal cells that promote carcinogenesis and progression of various malignant neoplasms. CAFs act through the secretion of metalloproteinases, growth factors, cytokines, mitochondrial DNA, and non-coding RNAs, among other molecules. Over the last few years, the evidence on the leading role of CAFs in gynecological cancers has notably increased, placing them as the cornerstone of neoplastic processes. In this review, the recently reported findings regarding the promoting role that CAFs play in gynecological cancers, their potential use as therapeutic targets, and the new evidence suggesting that they could act as tumor suppressors are analyzed and discussed.
Collapse
Affiliation(s)
- Julio César Villegas-Pineda
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Adrián Ramírez-de-Arellano
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Lesly Jazmín Bueno-Urquiza
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | | | - Ana Laura Pereira-Suárez
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- *Correspondence: Ana Laura Pereira-Suárez,
| |
Collapse
|
8
|
Malkova AM, Sharoyko VV, Zhukova NV, Gubal AR, Orlova RV. Laboratory biomarkers of an effective antitumor immune response. Clinical significance. Cancer Treat Res Commun 2021; 29:100489. [PMID: 34837797 DOI: 10.1016/j.ctarc.2021.100489] [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: 05/30/2021] [Revised: 10/18/2021] [Accepted: 11/09/2021] [Indexed: 10/19/2022]
Abstract
The modern checkpoint inhibitors block the programmed death-1 receptor and its ligand, cytotoxic T-lymphocyte-associated antigen 4 on tumor cells and lymphocytes, that induces cytotoxic reactions. Nowadays, there are no approved clinical and laboratory predictor markers of immune therapy efficacy, which would allow a more personalized approach to patient selection and treatment. The aim of this review is to analyze possible biomarkers of efficacy for treatment with checkpoint inhibitors according to the pathogenic mechanisms of drug action. The review revealed possible predictive biomarkers, that could be classified to 3 groups: biomarkers of high mutagenic potential of the tumor, biomarkers of high activity of adaptive immunity, biomarkers of low activity of the tumor microenvironment. The determination of the described markers before the start of therapy can be used to formulate a treatment regimen, in which the use of various immunomodulatory drugs, inhibitors of proinflammatory cytokines, angiogenic molecules, and probiotics can be considered.
Collapse
Affiliation(s)
- A M Malkova
- Saint Petersburg State University, 7/9 Universitetskaya Emb., St Petersburg 199034, Russian Federation.
| | - V V Sharoyko
- Saint Petersburg State University, 7/9 Universitetskaya Emb., St Petersburg 199034, Russian Federation.
| | - N V Zhukova
- Saint Petersburg State University, 7/9 Universitetskaya Emb., St Petersburg 199034, Russian Federation.
| | - A R Gubal
- Saint Petersburg State University, 7/9 Universitetskaya Emb., St Petersburg 199034, Russian Federation.
| | - R V Orlova
- Saint Petersburg State University, 7/9 Universitetskaya Emb., St Petersburg 199034, Russian Federation.
| |
Collapse
|
9
|
The Newly Synthetized Chalcone L1 Is Involved in the Cell Growth Inhibition, Induction of Apoptosis and Suppression of Epithelial-to-Mesenchymal Transition of HeLa Cells. Molecules 2021; 26:molecules26051356. [PMID: 33802621 PMCID: PMC7961543 DOI: 10.3390/molecules26051356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/24/2021] [Accepted: 02/28/2021] [Indexed: 12/24/2022] Open
Abstract
Over the past decades, natural products have emerged as promising agents with multiple biological activities. Many studies suggest the antioxidant, antiangiogenic, antiproliferative and anticancer effects of chalcones and their derivatives. Based on these findings, we decided to evaluate the effects of the newly synthetized chalcone L1 in a human cervical carcinoma cell (HeLa) model. Presented results were obtained by western blot and flow cytometric analyses, live cell imaging and antimigratory potential of L1 in HeLa cells was demonstrated by scratch assay. In the present study, we proved the role of L1 as an effective agent with antiproliferative activity supported by G2/M cell cycle arrest and apoptosis. Moreover, we proved that L1 is involved in modulating Transforming Growth Factor-β1 (TGF-β) signal transduction through Smad proteins and it also modulates other signalling pathways including Akt, JNK, p38 MAPK, and Erk1/2. The involvement of L1 in epithelial-to-mesenchymal transition was demonstrated by the regulation of N-cadherin, E-cadherin, and MMP-9 levels. Here, we also evaluated the effect of conditioned medium from BJ-5ta human foreskin fibroblasts in HeLa cell cultures with subsequent L1 treatment. Taken together, these data suggest the potential role of newly synthesized chalcone L1 as an anticancer-tumour microenvironment modulating agent.
Collapse
|
10
|
Chen Q, Hu L, Huang D, Chen K, Qiu X, Qiu B. Six-lncRNA Immune Prognostic Signature for Cervical Cancer. Front Genet 2020; 11:533628. [PMID: 33173530 PMCID: PMC7591729 DOI: 10.3389/fgene.2020.533628] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Background This study searched for immune-related long noncoding RNAs (lncRNAs) to predict the prognosis of patients with cervical cancer. Method We obtained immunologically relevant lncRNA expression profiles and clinical follow-up data from cervical cancer patients from The Cancer Genome Atlas database and the Molecular Signatures Database. Cervical cancer patients were randomly divided into a training group, testing group and combined group. The immune prognostic signature was constructed by Least Absolute Shrinkage and Selection Operator Cox regression, prognosis was analyzed by Kaplan-Meier curves between different groups, and the accuracy of the prognostic model was assessed by receiver operating characteristic-area under the curve (ROC-AUC) analysis. Results A six-lncRNA immune prognostic signature (LIPS) was constructed to predict the prognosis of cervical cancer. The six lncRNAs are as follows: AC009065.8, LINC01871, MIR210HG, GEMIN7-AS1, GAS5-AS1, and DLEU1. A ROC-AUC analysis indicated that the model could predict the prognosis of cervical cancer patients in different subgroups. A Kaplan-Meier analysis showed that patients with high risk scores had a poor prognosis; these results were equally meaningful in the subgroup analyses. Risk scores differed depending on the clinical pathology and tumor grade and were independent risk factors for cervical cancer prognosis. Gene set enrichment analysis revealed an association between the LIPS and the immune response, Wnt signaling pathway, and TGF beta signaling pathway. Conclusion Our study shows that the six-LIPS can predict the prognosis of cervical cancer and contribute to decisions regarding the immunotherapeutic strategy.
Collapse
Affiliation(s)
- Qian Chen
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Lang Hu
- Guangxi Medical University Cancer Hospital, Nanning, China
| | - Dongping Huang
- Department of Nutrition, School of Public Health, Guangxi Medical University, Nanning, China
| | - Kaihua Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaoqiang Qiu
- Department of Epidemiology, School of Public Health, Guangxi Medical University, Nanning, China
| | - Bingqing Qiu
- Department of Nuclear Medicine, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
11
|
Teixeira AF, Ten Dijke P, Zhu HJ. On-Target Anti-TGF-β Therapies Are Not Succeeding in Clinical Cancer Treatments: What Are Remaining Challenges? Front Cell Dev Biol 2020; 8:605. [PMID: 32733895 PMCID: PMC7360684 DOI: 10.3389/fcell.2020.00605] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 06/19/2020] [Indexed: 12/24/2022] Open
Abstract
Metastasis is the leading cause of death for cancer patients. During cancer progression, the initial detachment of cells from the primary tumor and the later colonization of a secondary organ are characterized as limiting steps for metastasis. Epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) are opposite dynamic multistep processes that enable these critical events in metastasis by altering the phenotype of cancer cells and improving their ability to migrate, invade and seed at distant organs. Among the molecular pathways that promote tumorigenesis in late-stage cancers, transforming growth factor-β (TGF-β) is described as an EMT master inducer by controlling different genes and proteins related to cytoskeleton assembly, cell-cell attachment and extracellular matrix remodeling. Still, despite the successful outcomes of different TGF-β pharmacological inhibitors in cell culture (in vitro) and animal models (in vivo), results in cancer clinical trials are poor or inconsistent at least, highlighting the existence of crucial components in human cancers that have not been properly explored. Here we review most recent findings to provide perspectives bridging the gap between on-target anti-TGF-β therapies in vitro and in pre-clinical models and the poor clinical outcomes in treating cancer patients. Specifically, we focus on (i) the dual roles of TGF-β signaling in cancer metastasis; (ii) dynamic signaling; (iii) functional differences of TGF-β free in solution vs. in exosomes; (iv) the regulatory effects of tumor microenvironment (TME) – particularly by cancer-associated fibroblasts – on TGF-β signaling pathway. Clearly identifying and establishing those missing links may provide strategies to revitalize and clinically improve the efficacy of TGF-β targeted therapies.
Collapse
Affiliation(s)
- Adilson Fonseca Teixeira
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Peter Ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Hong-Jian Zhu
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
12
|
Tong Y, Yang L, Yu C, Zhu W, Zhou X, Xiong Y, Wang W, Ji F, He D, Cao X. Tumor-Secreted Exosomal lncRNA POU3F3 Promotes Cisplatin Resistance in ESCC by Inducing Fibroblast Differentiation into CAFs. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:1-13. [PMID: 32637576 PMCID: PMC7321817 DOI: 10.1016/j.omto.2020.05.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 05/27/2020] [Indexed: 12/30/2022]
Abstract
Cancer-associated fibroblasts (CAFs), an activated subpopulation of fibroblasts, occupy a central position in the tumor microenvironment and have been shown to promote chemoresistance in multiple cancer types by secreting inflammatory cytokines. Herein, we report that tumor-secreted exosomal long non-coding RNAs (lncRNAs) can regulate cisplatin resistance in esophageal squamous cell carcinoma (ESCC) through transformation of normal fibroblasts (NFs) to CAFs. Primary CAFs and matched NFs were isolated from tumor tissues and matched normal esophageal epithelial tissues of ESCC patients. Fluorescence microscopy and qRT-PCR were used to investigate the transportation of exosomal lncRNAs from ESCC cells to NFs. To identify the specific lncRNAs involved, 14 ESCC-related lncRNAs were measured in NFs after incubation with exosomes from ESCC cells. We demonstrated that lncRNA POU3F3 can be transferred from ESCC cells to NFs via exosomes and that it mediated fibroblast activation. Activated fibroblasts further promoted proliferation and cisplatin resistance of ESCC cells through secreting interleukin 6 (IL-6). Moreover, our clinical data showed that high levels of plasma exosomal lncRNA POU3F3 correlated significantly with lack of complete response and poor survival in ESCC patients. Therefore, these data demonstrate that lncRNA POU3F3 is involved in cisplatin resistance in ESCC and that this effect is mediated through exosomal lncRNA POU3F3-induced transformation of NFs to CAFs.
Collapse
Affiliation(s)
- Yusuo Tong
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Lili Yang
- Department of Oncology Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Changhua Yu
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Weiguo Zhu
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xilei Zhou
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Yaozu Xiong
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Wanwei Wang
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Fuzhi Ji
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Dongcheng He
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xiufeng Cao
- Department of Oncology Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Thoracic Surgery, Taikang Xianlin Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
13
|
Teixeira AF, Ten Dijke P, Zhu HJ. On-Target Anti-TGF-β Therapies Are Not Succeeding in Clinical Cancer Treatments: What Are Remaining Challenges? Front Cell Dev Biol 2020. [PMID: 32733895 DOI: 10.3389/fcell.2020.00605.pmid:32733895;pmcid:pmc7360684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Metastasis is the leading cause of death for cancer patients. During cancer progression, the initial detachment of cells from the primary tumor and the later colonization of a secondary organ are characterized as limiting steps for metastasis. Epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) are opposite dynamic multistep processes that enable these critical events in metastasis by altering the phenotype of cancer cells and improving their ability to migrate, invade and seed at distant organs. Among the molecular pathways that promote tumorigenesis in late-stage cancers, transforming growth factor-β (TGF-β) is described as an EMT master inducer by controlling different genes and proteins related to cytoskeleton assembly, cell-cell attachment and extracellular matrix remodeling. Still, despite the successful outcomes of different TGF-β pharmacological inhibitors in cell culture (in vitro) and animal models (in vivo), results in cancer clinical trials are poor or inconsistent at least, highlighting the existence of crucial components in human cancers that have not been properly explored. Here we review most recent findings to provide perspectives bridging the gap between on-target anti-TGF-β therapies in vitro and in pre-clinical models and the poor clinical outcomes in treating cancer patients. Specifically, we focus on (i) the dual roles of TGF-β signaling in cancer metastasis; (ii) dynamic signaling; (iii) functional differences of TGF-β free in solution vs. in exosomes; (iv) the regulatory effects of tumor microenvironment (TME) - particularly by cancer-associated fibroblasts - on TGF-β signaling pathway. Clearly identifying and establishing those missing links may provide strategies to revitalize and clinically improve the efficacy of TGF-β targeted therapies.
Collapse
Affiliation(s)
- Adilson Fonseca Teixeira
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Peter Ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Hong-Jian Zhu
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
14
|
Cheng L, Shi X, Huo D, Zhao Y, Zhang H. MiR-449b-5p regulates cell proliferation, migration and radioresistance in cervical cancer by interacting with the transcription suppressor FOXP1. Eur J Pharmacol 2019; 856:172399. [PMID: 31103631 DOI: 10.1016/j.ejphar.2019.05.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 01/13/2023]
Abstract
MicroRNAs (miRNAs) have been acknowledged as crucial regulators for the malignant progression of human cancers. However, the molecular mechanism associated with the malignant progression of cervical cancer (CC) is still largely unmarked. The focus of this study is to explore the potential functional mechanism of miR-449b-5p in CC. Using qRT-PCR analysis, we detected a relative lower expression level of miR-449b-5p in CC tissues and cell lines by comparing with the normal tissues and cells. Low level of miR-449b-5p in CC cell lines was further demonstrated by northern blot. Subsequently, downregulation of miR-449b-5p was closely correlated with the low overall survival rate of patients with cervical cancer. Gain-of-function assays demonstrated that upregulation of miR-449b-5p had an inhibitory effect on cell proliferation, migration and invasion. Moreover, FOXP1 was found to be a transcription suppressor and downstream target of miR-449b-5p. Rescue assays and in vivo experiment were applied to demonstrate the biological function of miR-449b-5p-FOXP1 feedback loop. In summary, miR-449b-5p interacted with FOXP1 to regulate cell proliferation, migration, invasion and radiosensitivity in CC.
Collapse
Affiliation(s)
- Lei Cheng
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, 266035, PR China
| | - Xingzhen Shi
- Department of Obstetrics and Gynecology, The First People's Hospital of Jinan, Jinan, Shandong, 250011, PR China
| | - Demei Huo
- Department of Obstetrics and Gynecology, The First People's Hospital of Jinan, Jinan, Shandong, 250011, PR China
| | - Ying Zhao
- Department of Anesthesiology, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, 266035, PR China
| | - Hui Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
15
|
Deckey DG, Reid DBC, Mallozzi S, Daniels AH. Cutaneous Metastasis via Surgical Planes After Lumbar Spinal Reconstruction for Spinal Metastatic Disease. World Neurosurg 2018; 123:49-53. [PMID: 30528523 DOI: 10.1016/j.wneu.2018.11.218] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Cervical squamous cell carcinoma is the second most common cancer in women worldwide and the third most common cause of female cancer mortality. It frequently metastasizes to the osseous spine and has been rarely reported to cause cutaneous metastases at gynecologic surgical scars. CASE DESCRIPTION A 63-year-old woman with known metastatic cervical squamous cell carcinoma presented with a pathologic fracture of the L2 vertebra, unrelenting back pain, bilateral lower extremity weakness, and inability to ambulate. The patient elected to undergo an L2 corpectomy with intervertebral cage placement via the right-sided retroperitoneal approach followed by percutaneous L1-L4 pedicle screw instrumentation in the same operative setting. Her immediate postoperative course was uneventful. Eight weeks after surgery, both her flank and posterior incisions became painful and swollen. She underwent irrigation and debridement of the right flank incision due to elevated inflammatory markers and concern for surgical site infection. Multiple intraoperative cultures were obtained and were ultimately negative for bacterial or fungal growth. Several weeks later, the patient presented with worsening painful growths of her incisions. Excisional biopsy and debulking of the masses in the operating room revealed squamous cell carcinoma with areas of cystic change and tumor necrosis consistent with progression of the patient's metastatic cervical cancer. CONCLUSIONS Cervical squamous cell carcinoma can metastasize regionally via surgical planes and manifest as cutaneous lesions. Local metastasis should be included in the differential in cases of postoperative wound complications.
Collapse
Affiliation(s)
- David G Deckey
- Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Daniel B C Reid
- Department of Orthopaedics, Alpert Medical School of Brown University, East Providence, Rhode Island, USA
| | - Scott Mallozzi
- Department of Orthopaedics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Alan H Daniels
- Division of Spine Surgery, Department of Orthopaedics, Alpert Medical School of Brown University, East Providence, Rhode Island, USA.
| |
Collapse
|
16
|
Negative roles of B7-H3 and B7-H4 in the microenvironment of cervical cancer. Exp Cell Res 2018; 371:222-230. [PMID: 30099052 DOI: 10.1016/j.yexcr.2018.08.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 08/08/2018] [Indexed: 01/18/2023]
Abstract
Although persistent human papilloma virus (HPV) infection exerts a crucial influence on cervical carcinogenesis, other factors are also involved in its development, such as intraepithelial lesions and cervical cancer. B7-H3 and B7-H4, which have been reported to be co-regulatory ligands in the B7 family, had been found to be overexpressed in cervical cancer and correlated with adverse clinicopathological features and poor prognosis in our previous studies. In this study, we sought to explore the effects of B7-H3 and B7-H4 on the cervical microenvironment. Among several immune cytokines, interleukin-10 (IL-10) and transforming growth factor (TGF) β1 stand out as important immunosuppressive factors. Our studies found that IL-10 expression increased with pathological change levels and significantly correlated with cervical cancer differentiation (P < 0.05). TGF-β1 correlated with lymph node metastasis (LNM) (P < 0.01). Expression of B7-H3 and B7-H4 positively correlated with the expression of IL-10 and TGF-β1. After co-culture, we found that overexpression of B7-H3 and B7-H4 in cervical cancer cell lines resulted in activation of the cell cycle and decreased apoptosis of U-937 cells. In addition, the contents of IL-10 and TGF-β1, as well as their protein expression levels, increased in co-culture supernatants in U-937 cells, suggesting regulation by the p-JAK2/STAT3 pathway. The in vivo results demonstrated that with the increasing expression of B7-H3/B7-H4, the expression of IL-10 and TGF-β1 also increased significantly. Overall, the expression of B7-H3 and B7-H4 favored an immunosuppressive microenvironment by promoting the production of IL-10 and TGF-β1, thereby resulting in progression of cervical carcinogenesis.
Collapse
|
17
|
Wei WF, Zhou CF, Wu XG, He LN, Wu LF, Chen XJ, Yan RM, Zhong M, Yu YH, Liang L, Wang W. MicroRNA-221-3p, a TWIST2 target, promotes cervical cancer metastasis by directly targeting THBS2. Cell Death Dis 2017; 8:3220. [PMID: 29242498 PMCID: PMC5870596 DOI: 10.1038/s41419-017-0077-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/26/2017] [Accepted: 09/27/2017] [Indexed: 01/05/2023]
Abstract
MicroRNAs have implicated in the relapse and metastasis of cervical cancer, which is the leading cause of cervical cancer-related mortality. However, the underlying molecular mechanisms need further elucidation. Our present study revealed that miR-221-3p is transcriptionally promoted in metastatic cervical cancer tissues compared with non-metastatic cervical cancer tissues. Forced overexpression of miR-221-3p facilitated EMT and promoted cell migration and invasion in vitro and lymphatic metastasis in vivo. Twist homolog 2 (TWIST2) was found to be a key transcription factor binding to the promoter of miR-221-3p. Inhibitors of miR-221-3p drastically reduced the induction of EMT and decreased cell migration and invasion mediated by TWIST2. By combined computational and experimental approaches, THBS2 was recognized to be an important downstream target gene of miR-221-3p. In cervical cancer tissues, especially with lymphatic metastasis, miR-221-3p and TWIST2 were increased and THBS2 was decreased, suggesting that TWIST2 induces miR-221-3p expression and consequently suppresses its direct target THBS2 in lymphatic metastasis CC. Our findings uncover a mechanistic role for miR-221-3p in lymph node metastasis, suggesting that miR-221-3p is upregulated by the transcription factor TWIST2 and downregulates its target THBS2, which may potentially promote lymph node metastasis in cervical cancer.
Collapse
Affiliation(s)
- Wen-Fei Wei
- Department of Obstetrics and Gynecology, Nanfang Hospital/First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Chen-Fei Zhou
- Department of Obstetrics and Gynecology, Nanfang Hospital/First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Xiang-Guang Wu
- Department of Obstetrics and Gynecology, Nanfang Hospital/First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Li-Na He
- Department of Obstetrics and Gynecology, Nanfang Hospital/First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Lan-Fang Wu
- Department of Obstetrics and Gynecology, Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Xiao-Jing Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital/First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Rui-Ming Yan
- Department of Obstetrics and Gynecology, Nanfang Hospital/First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital/First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Yan-Hong Yu
- Department of Obstetrics and Gynecology, Nanfang Hospital/First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Li Liang
- Department of pathology, Nanfang Hospital/First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.
| | - Wei Wang
- Department of Obstetrics and Gynecology, Nanfang Hospital/First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China.
| |
Collapse
|
18
|
Woodby B, Scott M, Bodily J. The Interaction Between Human Papillomaviruses and the Stromal Microenvironment. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:169-238. [PMID: 27865458 PMCID: PMC5727914 DOI: 10.1016/bs.pmbts.2016.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human papillomaviruses (HPVs) are small, double-stranded DNA viruses that replicate in stratified squamous epithelia and cause a variety of malignancies. Current efforts in HPV biology are focused on understanding the virus-host interactions that enable HPV to persist for years or decades in the tissue. The importance of interactions between tumor cells and the stromal microenvironment has become increasingly apparent in recent years, but how stromal interactions impact the normal, benign life cycle of HPVs, or progression of lesions to cancer is less understood. Furthermore, how productively replicating HPV impacts cells in the stromal environment is also unclear. Here we bring together some of the relevant literature on keratinocyte-stromal interactions and their impacts on HPV biology, focusing on stromal fibroblasts, immune cells, and endothelial cells. We discuss how HPV oncogenes in infected cells manipulate other cells in their environment, and, conversely, how neighboring cells may impact the efficiency or course of HPV infection.
Collapse
Affiliation(s)
- B Woodby
- Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - M Scott
- Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - J Bodily
- Louisiana State University Health Sciences Center, Shreveport, LA, United States.
| |
Collapse
|
19
|
CAF cellular glycolysis: linking cancer cells with the microenvironment. Tumour Biol 2016; 37:8503-14. [PMID: 27075473 DOI: 10.1007/s13277-016-5049-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/01/2016] [Indexed: 02/06/2023] Open
Abstract
Cancers have long being hallmarked as cells relying heavily on their glycolysis for energy generation in spite of having functional mitochondria. The metabolic status of the cancer cells have been revisited time and again to get better insight into the overall carcinogenesis process which revealed the apparent crosstalks between the cancer cells with the fibroblasts present in the tumour microenvironment. This review focuses on the mechanisms of transformations of normal fibroblasts to cancer-associated fibroblasts (CAF), the participation of the CAF in tumour progression with special interest to the role of CAF cellular glycolysis in the overall tumorigenesis. The fibroblasts, when undergoes the transformation process, distinctly switches to a more glycolytic phenotype in order to provide the metabolic intermediates necessary for carrying out the mitochondrial pathways of ATP generation in cancer cells. This review will also discuss the molecular mechanisms responsible for this metabolic make over promoting glycolysis in CAF cells. A thorough investigation of the pathways and molecules involved will not only help in understanding the process of activation and metabolic reprogramming in CAF cells but also might open up new targets for cancer therapy.
Collapse
|
20
|
Zhu H, Luo H, Shen Z, Hu X, Sun L, Zhu X. Transforming growth factor-β1 in carcinogenesis, progression, and therapy in cervical cancer. Tumour Biol 2016; 37:7075-83. [PMID: 27010470 DOI: 10.1007/s13277-016-5028-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 03/18/2016] [Indexed: 01/20/2023] Open
Abstract
Transforming growth factor β1 (TGF-β1) is a multifunctional cytokine that plays important roles in cervical tumor formation, invasion, progression, and metastasis. TGF-β1 functions as a tumor inhibitor in precancerous lesions and early stage cancers of cervix whereas as a tumor promoter in later stage. This switch from a tumor inhibitor to a tumor promoter might be due to various alterations in TGF-β signaling pathway, such as mutations or loss of expression of TGF-β receptors and SMAD proteins. Additionally, the oncoproteins of human papillomaviruses have been shown to stimulate TGF-β1 expression, which in turn suppresses host immune surveillance. Thus, in addition to driving tumor cell migration and metastasis, TGF-β1 is believed to play a key role in promoting human papillomavirus infection by weakening host immune defense. In this article, we will discuss the role of TGF-β1 in the expression, carcinogenesis, progression, and therapy in cervical cancers. A better understanding of this cytokine in cervical carcinogenesis is essential for critical evaluation of this cytokine as a potential prognostic marker and therapeutic target.
Collapse
Affiliation(s)
- Haiyan Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, China
| | - Hui Luo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, China
| | - Zhaojun Shen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, China
| | - Xiaoli Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, China
| | - Luzhe Sun
- Department of Cellular & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, 325027, China.
| |
Collapse
|
21
|
MiR-519d facilitates the progression and metastasis of cervical cancer through direct targeting Smad7. Cancer Cell Int 2016; 16:21. [PMID: 27006642 PMCID: PMC4802873 DOI: 10.1186/s12935-016-0298-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/11/2016] [Indexed: 12/19/2022] Open
Abstract
Background MicroRNAs (miRNAs) play pivotal roles in the development of various cancer types, including cervical cancer. Methods and results In this study, we showed that miR-519d, a miRNA within the chromosome 19 miRNA cluster, was significantly upregulated in cervical cancer tissues, compared with non-tumorous cervical samples. Suppression of miR-519d markedly attenuated the migration and invasion of HeLa and SiHa cervical cancer cells. Additionally, miR-519d inhibited the apoptosis of cervical cancer cells, and the proliferation of cervical cancer cells was also affected following transfection of miR-519d inhibitor. Moreover, we identified Smad7 to be a novel target of miR-519d in cervical cancer cells. MiR-519d matched the 3′-UTR of Smad7 mRNA. Transfection with miR-519d mimics led to apparent downregulation of Smad7 both at the mRNA and protein levels. Luciferase reporter analysis revealed that miR-519d reduced the luciferase activity of Smad7 mRNA 3′-UTR through matching site-dependent manner. And more notably, suppression of Smad7 remarkably restored the migration and invasion of miR-519d-depleted cervical cancer cells. Conclusion Taken together, these findings implicated that miR-519d promoted the progression and metastasis of cervical cancer through targeting Smad7.
Collapse
|
22
|
Cheng Y, Guo Y, Zhang Y, You K, Li Z, Geng L. MicroRNA-106b is involved in transforming growth factor β1-induced cell migration by targeting disabled homolog 2 in cervical carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:11. [PMID: 26769181 PMCID: PMC4714510 DOI: 10.1186/s13046-016-0290-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 01/10/2016] [Indexed: 11/10/2022]
Abstract
BACKGROUND MicroRNA-106b (miR-106b) was recently identified as an oncogene participating in cancer progression. Transforming growth factor β1(TGF-β1) is an indispensable cytokine regulating the local microenvironment, thereby promoting cervical cancer progression. However, the roles of miR-106b in cervical carcinoma progression and TGF-β1-involvement in the tumorigenesis of cervical cancer remain unknown. METHODS The expression of miR-106b in human cervical specimens was detected by real-time PCR analysis and in situ hybridization assay. The effect of miR-106b on cell migration was analyzed by scratch and transwell assays. TGF-β1 was used to induce cell migration. The expression of the miR-106b target gene DAB2 in human cervical tissues and cell lines were measured by qRT-PCR, western blot and immunohistochemistry. Dual-luciferase reporter assay was used to identify DAB2 as a miR-106b-directed target gene. RESULTS miR-106b was frequently up-regulated in human cervical carcinoma specimens and cervical cancer cell lines. Over-expression of miR-106b significantly promoted HeLa and SiHa cells migration. Likewise, inhibition of miR-106b decreased HeLa and SiHa cells migration. The multifunctional cytokine TGF-β facilitates metastasis in cervical carcinoma. miR-106b inhibitor treatment decreased the TGF-β1-stimulated migration of HeLa and SiHa cells. DAB2, a predicted target gene of miR-106b, was inhibited by TGF-β1 partly through miR-106b and was involved in TGF-β1-induced cervical cancer cell migration. The expression of DAB2 was low in cervical cancer tissues, and negatively correlated with miR-106b expression. Finally, DAB2 was identified as a miR-106b-directed target gene by dual-luciferase reporter assay. CONCLUSION Our data suggest that the TGF-β1/miR-106b/DAB2 axis may be involved in the pathogenesis of cervical carcinoma.
Collapse
Affiliation(s)
- Yuan Cheng
- Department of Gynecology and Obstetrics, Peking University Third Hospital, Beijing, 100191, China.
| | - Yanli Guo
- Department of Gynecology and Obstetrics, Peking University Third Hospital, Beijing, 100191, China.
| | - Youyi Zhang
- Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovasicular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China.
| | - Ke You
- Department of Gynecology and Obstetrics, Peking University Third Hospital, Beijing, 100191, China.
| | - Zijian Li
- Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovasicular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China.
| | - Li Geng
- Department of Gynecology and Obstetrics, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|