1
|
Chernosky NM, Tamagno I, Polak KL, Chan ER, Yuan X, Jackson MW. Toll-Like receptor 3-mediated interferon-β production is suppressed by oncostatin m and a broader epithelial-mesenchymal transition program. Breast Cancer Res 2024; 26:167. [PMID: 39593161 PMCID: PMC11590466 DOI: 10.1186/s13058-024-01918-2] [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: 06/17/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Patients with Triple Negative Breast Cancer (TNBC) currently lack targeted therapies, and consequently face higher mortality rates when compared to patients with other breast cancer subtypes. The tumor microenvironment (TME) cytokine Oncostatin M (OSM) reprograms TNBC cells to a more stem-like/mesenchymal state, conferring aggressive cancer cell properties such as enhanced migration and invasion, increased tumor-initiating capacity, and intrinsic resistance to the current standards of care. In contrast to OSM, Interferon-β (IFN-β) promotes a more differentiated, epithelial cell phenotype in addition to its role as an activator of anti-tumor immunity. Importantly, OSM suppresses the production of IFN-β, although the mechanism of IFN-β suppression has not yet been elucidated. METHODS IFN-β production and downstream autocrine signaling were assessed via quantitative real-time PCR (qRT-PCR) and Western blotting in TNBC cells following exposure to OSM. RNA-sequencing (RNA-seq) was used to assess an IFN-β metagene signature, and to assess the expression of innate immune sensors, which are upstream activators of IFN-β. Cell migration was assessed using an in vitro chemotaxis assay. Additionally, TNBC cells were exposed to TGF-β1, Snail, and Zeb1, and IFN-β production and downstream autocrine signaling were assessed via RNA-seq, qRT-PCR, and Western blotting. RESULTS Here, we identify the repression of Toll-like Receptor 3 (TLR3), an innate immune sensor, as the key molecular event linking OSM signaling and the repression of IFN-β transcription, production, and autocrine IFN signaling. Moreover, we demonstrate that additional epithelial-mesenchymal transition-inducing factors, such as TGF-β1, Snail, and Zeb1, similarly suppress TLR3-mediated IFN-β production and signaling. CONCLUSIONS Our findings provide a novel insight into the regulation of TLR3 and IFN-β production in TNBC cells, which are known indicators of treatment responses to DNA-damaging therapies. Furthermore, strategies to stimulate TLR3 in order to increase IFN-β within the TME may be ineffective in stem-like/mesenchymal cells, as TLR3 is strongly repressed. Rather, we propose that therapies targeting OSM or OSM receptor would reverse the stem-like/mesenchymal program and restore TLR3-mediated IFN-β production within the TME, facilitating improved responses to current therapies.
Collapse
Affiliation(s)
- Noah M Chernosky
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - Ilaria Tamagno
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - Kelsey L Polak
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - E Ricky Chan
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
- Cleveland Institute for Computational Biology, Cleveland, OH, 44106, USA
| | - Xueer Yuan
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - Mark W Jackson
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
2
|
Polak KL, Tamagno I, Parameswaran N, Smigiel J, Chan ER, Yuan X, Rios B, Jackson MW. Oncostatin-M and OSM-Receptor Feed-Forward Activation of MAPK Induces Separable Stem-like and Mesenchymal Programs. Mol Cancer Res 2023; 21:975-990. [PMID: 37310811 PMCID: PMC10527478 DOI: 10.1158/1541-7786.mcr-22-0715] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 04/19/2023] [Accepted: 06/08/2023] [Indexed: 06/15/2023]
Abstract
Patients diagnosed with pancreatic ductal adenocarcinoma (PDAC) frequently present with advanced metastatic disease and exhibit a poor response to therapy, resulting in poor outcomes. The tumor microenvironment cytokine Oncostatin-M (OSM) initiates PDAC plasticity, inducing the reprogramming to a stem-like/mesenchymal state, which enhances metastasis and therapy resistance. Using a panel of PDAC cells driven through epithelial-mesenchymal transition (EMT) by OSM or the transcription factors ZEB1 or SNAI1, we find that OSM uniquely induces tumor initiation and gemcitabine resistance independently of its ability to induce a CD44HI/mesenchymal phenotype. In contrast, while ZEB1 and SNAI1 induce a CD44HI/mesenchymal phenotype and migration comparable with OSM, they are unable to promote tumor initiation or robust gemcitabine resistance. Transcriptomic analysis identified that OSM-mediated stemness requires MAPK activation and sustained, feed-forward transcription of OSMR. MEK and ERK inhibitors prevented OSM-driven transcription of select target genes and stem-like/mesenchymal reprogramming, resulting in reduced tumor growth and resensitization to gemcitabine. We propose that the unique properties of OSMR, which hyperactivates MAPK signaling when compared with other IL6 family receptors, make it an attractive therapeutic target, and that disrupting the OSM-OSMR-MAPK feed-forward loop may be a novel way to therapeutically target the stem-like behaviors common to aggressive PDAC. IMPLICATIONS Small-molecule MAPK inhibitors may effectively target the OSM/OSMR-axis that leads to EMT and tumor initiating properties that promote aggressive PDAC.
Collapse
Affiliation(s)
- Kelsey L Polak
- Department of Pathology and Case Comprehensive Cancer Center, Case Western Reserve University Cleveland, OH, USA
| | - Ilaria Tamagno
- Department of Pathology and Case Comprehensive Cancer Center, Case Western Reserve University Cleveland, OH, USA
| | - Neetha Parameswaran
- Department of Pathology and Case Comprehensive Cancer Center, Case Western Reserve University Cleveland, OH, USA
| | - Jacob Smigiel
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - E. Ricky Chan
- Department of Pathology and Case Comprehensive Cancer Center, Case Western Reserve University Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Xueer Yuan
- Department of Pathology and Case Comprehensive Cancer Center, Case Western Reserve University Cleveland, OH, USA
| | - Brenda Rios
- Cancer Biology Program, Vanderbilt School of Medicine, Nashville, Tennessee, USA
| | - Mark W. Jackson
- Department of Pathology and Case Comprehensive Cancer Center, Case Western Reserve University Cleveland, OH, USA
| |
Collapse
|
3
|
Lin J, Ye S, Ke H, Lin L, Wu X, Guo M, Jiao B, Chen C, Zhao L. Changes in the mammary gland during aging and its links with breast diseases. Acta Biochim Biophys Sin (Shanghai) 2023. [PMID: 37184281 DOI: 10.3724/abbs.2023073] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
The functional capacity of organisms declines in the process of aging. In the case of breast tissue, abnormal mammary gland development can lead to dysfunction in milk secretion, a primary function, as well as the onset of various diseases, such as breast cancer. In the process of aging, the terminal duct lobular units (TDLUs) within the breast undergo gradual degeneration, while the proportion of adipose tissue in the breast continues to increase and hormonal levels in the breast change accordingly. Here, we review changes in morphology, internal structure, and cellular composition that occur in the mammary gland during aging. We also explore the emerging mechanisms of breast aging and the relationship between changes during aging and breast-related diseases, as well as potential interventions for delaying mammary gland aging and preventing breast disease.
Collapse
Affiliation(s)
- Junqiang Lin
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Shihui Ye
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Hao Ke
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Liang Lin
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Xia Wu
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Mengfei Guo
- Huankui Academy, Nanchang University, Nanchang 330031, China
| | - Baowei Jiao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Ceshi Chen
- Academy of Biomedical Engineering, Kunming Medical University, Kunming 650500, China
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- the Third Affiliated Hospital, Kunming Medical University, Kunming 650118, China
| | - Limin Zhao
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| |
Collapse
|
4
|
Estepa-Fernández A, García-Fernández A, Lérida-Viso A, Blandez JF, Galiana I, Sancenon-Galarza F, Orzáez M, Martínez-Máñez R. Combination of palbociclib with navitoclax based-therapies enhances in vivo antitumoral activity in triple-negative breast cancer. Pharmacol Res 2023; 187:106628. [PMID: 36566002 DOI: 10.1016/j.phrs.2022.106628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 11/29/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Triple-negative breast cancer (TNBC) is a very aggressive subtype of breast cancer with a poor prognosis and limited effective therapeutic options. Induction of senescence, arrest of cell proliferation, has been explored as an effective method to limit tumor progression in metastatic breast cancer. However, relapses occur in some patients, possibly as a result of the accumulation of senescent tumor cells in the body after treatment, which promote metastasis. In this study, we explored the combination of senescence induction and the subsequent removal of senescent cells (senolysis) as an alternative approach to improve outcomes in TNBC patients. We demonstrate that a combination treatment, using the senescence-inducer palbociclib and the senolytic agent navitoclax, delays tumor growth and reduces metastases in a mouse xenograft model of aggressive human TNBC (hTNBC). Furthermore, considering the off-target effects and toxicity derived from the use of navitoclax, we propose a strategy aimed at minimizing the associated side effects. We use a galacto-conjugated navitoclax (nav-Gal) as a senolytic prodrug that can preferentially be activated by β-galactosidase overexpressed in senescent cells. Concomitant treatment with palbociclib and nav-Gal in vivo results in the eradication of senescent hTNBC cells with consequent reduction of tumor growth, while reducing the cytotoxicity of navitoclax. Taken together, our results support the efficacy of combination therapy of senescence-induction with senolysis for hTNBC, as well as the development of a targeted approach as an effective and safer therapeutic opportunity.
Collapse
Affiliation(s)
- Alejandra Estepa-Fernández
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain
| | - Alba García-Fernández
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain.
| | - Araceli Lérida-Viso
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026 Valencia, Spain
| | - Juan F Blandez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026 Valencia, Spain
| | - Irene Galiana
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - Félix Sancenon-Galarza
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026 Valencia, Spain
| | - Mar Orzáez
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València. Camino de Vera, s/n, 46022 Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe. C/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026 Valencia, Spain.
| |
Collapse
|
5
|
Grubb T, Maganti S, Krill-Burger JM, Fraser C, Stransky L, Radivoyevitch T, Sarosiek KA, Vazquez F, Kaelin WG, Chakraborty AA. A Mesenchymal Tumor Cell State Confers Increased Dependency on the BCL-XL Antiapoptotic Protein in Kidney Cancer. Clin Cancer Res 2022; 28:4689-4701. [PMID: 35776130 PMCID: PMC9633392 DOI: 10.1158/1078-0432.ccr-22-0669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/07/2022] [Accepted: 06/28/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Advanced/metastatic forms of clear-cell renal cell carcinomas (ccRCC) have limited therapeutic options. Genome-wide genetic screens have identified cellular dependencies in many cancers. Using the Broad Institute/Novartis combined short hairpin RNA (shRNA) dataset, and cross-validation with the CRISPR/Cas9 DepMap (21Q3) dataset, we sought therapeutically actionable dependencies in kidney lineage cancers. EXPERIMENTAL DESIGN We identified preferential genetic dependencies in kidney cancer cells versus other lineages. BCL2L1, which encodes the BCL-XL antiapoptotic protein, scored as the top actionable dependency. We validated this finding using genetic and pharmacologic tools in a panel of ccRCC cell lines. Select BCL-XL-dependent (versus independent) cell lines were then transcriptionally profiled to identify biomarkers and mechanistic drivers of BCL-XL dependence. Cell-based studies (in vitro and in vivo) and clinical validations were used to address physiologic relevance. RESULTS Inactivation of BCL-XL, but not BCL-2, led to fitness defects in renal cancer cells, and sensitized them to chemotherapeutics. Transcriptomic profiling identified a "BCL-XL dependency" signature, including an elevated mesenchymal gene signature. A mesenchymal state was both necessary and sufficient to confer increased BCL-XL dependence. The "BCL-XL dependency" signature was observed in approximately 30% of human ccRCCs, which were also associated with worse clinical outcomes. Finally, an orally bioavailable BCL-XL inhibitor, A-1331852, showed antitumor efficacy in vivo. CONCLUSIONS Our studies uncovered an unexpected link between cell state and BCL-XL dependence in ccRCC. Therapeutic agents that specifically target BCL-XL are available. Our work justifies testing the utility of BCL-XL blockade to target, likely, a clinically aggressive subset of human kidney cancers. See related commentary by Wang et al., p. 4600.
Collapse
Affiliation(s)
- Treg Grubb
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Smruthi Maganti
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Cameron Fraser
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.,Molecular and Integrative Physiology Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Laura Stransky
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Tomas Radivoyevitch
- Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kristopher A. Sarosiek
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.,Molecular and Integrative Physiology Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | | | - William G. Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.,Corresponding Authors: William G. Kaelin, Jr., Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave, Boston, MA 02115. . Phone: 617-632-3975; Abhishek A. Chakraborty, Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195. . Phone: 216-445-6620
| | - Abhishek A. Chakraborty
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.,Corresponding Authors: William G. Kaelin, Jr., Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave, Boston, MA 02115. . Phone: 617-632-3975; Abhishek A. Chakraborty, Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195. . Phone: 216-445-6620
| |
Collapse
|
6
|
The Role of the IL-6 Cytokine Family in Epithelial-Mesenchymal Plasticity in Cancer Progression. Int J Mol Sci 2021; 22:ijms22158334. [PMID: 34361105 PMCID: PMC8347315 DOI: 10.3390/ijms22158334] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/12/2021] [Accepted: 07/28/2021] [Indexed: 02/07/2023] Open
Abstract
Epithelial–mesenchymal plasticity (EMP) plays critical roles during embryonic development, wound repair, fibrosis, inflammation and cancer. During cancer progression, EMP results in heterogeneous and dynamic populations of cells with mixed epithelial and mesenchymal characteristics, which are required for local invasion and metastatic dissemination. Cancer development is associated with an inflammatory microenvironment characterized by the accumulation of multiple immune cells and pro-inflammatory mediators, such as cytokines and chemokines. Cytokines from the interleukin 6 (IL-6) family play fundamental roles in mediating tumour-promoting inflammation within the tumour microenvironment, and have been associated with chronic inflammation, autoimmunity, infectious diseases and cancer, where some members often act as diagnostic or prognostic biomarkers. All IL-6 family members signal through the Janus kinase (JAK)–signal transducer and activator of transcription (STAT) pathway and are able to activate a wide array of signalling pathways and transcription factors. In general, IL-6 cytokines activate EMP processes, fostering the acquisition of mesenchymal features in cancer cells. However, this effect may be highly context dependent. This review will summarise all the relevant literature related to all members of the IL-6 family and EMP, although it is mainly focused on IL-6 and oncostatin M (OSM), the family members that have been more extensively studied.
Collapse
|