1
|
Ren F, Pang X, Jin F, Luan N, Guo H, Zhu L. Integration of scRNA-seq and bulk RNA-seq to reveal the association and potential molecular mechanisms of metabolic reprogramming regulated by lactylation and chemotherapy resistance in ovarian cancer. Front Immunol 2025; 16:1513806. [PMID: 40093000 PMCID: PMC11907005 DOI: 10.3389/fimmu.2025.1513806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Objective Ovarian cancer (OC) ranks among the foremost causes of mortality in gynecological malignancies, with chemoresistance being the primary factor contributing to unfavorable prognosis. This work seeks to clarify the mechanisms of resistance-related lactylation in OC, intending to offer novel theoretical foundations and therapy strategies for addressing chemoresistance. Methods Through the combined analysis of bulk RNA-seq and single-cell RNA-seq data, we initially found lactylation genes linked to chemoresistance. Subsequently, we employed differential expression analysis, survival analysis, enrichment analysis, and other methodologies to further investigate the roles and molecular mechanisms of these genes in tumor resistance. Ultimately, we investigated the differential expression of these genes in resistant and non-resistant tissues and cells via experimentation. Results We found two candidate genes associated with lactylation chemoresistance, ALDH1A1 and S100A4. Analysis of single-cell data indicated that tumor cells represent the primary cell subpopulation relevant to resistance studies. Subpopulation analysis indicated that several tumor cell subtypes were markedly linked to resistance, with elevated expression levels of ALDH1A1 and S100A4 in the resistant subpopulation, notably correlating with various immunological and metabolic pathways. Analysis of metabolic pathways indicated that oxidative phosphorylation and glycolysis activity was elevated in the resistant subpopulation, and lactic acid buildup was associated with chemoresistance. The investigation of the marker gene protein-protein interaction network in the resistant subgroup elucidated the intricate interactions among these genes. The expression levels of ALDH1A1 and S100A4 in the OC tissues of the platinum-resistant cohort were markedly elevated compared to the sensitive cohort, with a considerable rise in S100A4 expression observed in resistant OC cells, demonstrating co-localization with lactylation. Conclusion This work elucidates the significant function of lactylation in OC chemoresistance and identifies ALDH1A1 and S100A4 as possible genes associated with drug resistance. These findings enhance our comprehension of the mechanisms behind chemoresistance in OC and offer critical insights for the formulation of novel therapeutic options.
Collapse
Affiliation(s)
| | | | | | | | | | - Liancheng Zhu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
2
|
Hayashi M, Yokoi A, Nakagawa M, Hashimura M, Oguri Y, Saegusa M. Prognostic Significance of S100A4 in Ovarian Clear Cell Carcinoma: Its Relation to Tumor Progression and Chemoresistance. Cancers (Basel) 2025; 17:184. [PMID: 39857966 PMCID: PMC11763377 DOI: 10.3390/cancers17020184] [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: 12/03/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES S100A4, a small calcium-binding protein, promotes metastasis in a variety of human malignancies, but little is known about its involvement in ovarian clear cell carcinoma (OCCC). Herein, we characterized the functional role of S100A4 in this tumor type. METHODS We analyzed immunohistochemical sections from 120 OCCC patients. OCCC cell lines in which S100A4 was knocked out (KO) or overexpressed were also used to study the protein's effects. RESULTS Stable overexpression of S100A4 decreased the proliferation of OCCC cell lines (concomitant with more cells in G1 and fewer in the G2/M phase of the cell cycle). S100A4 overexpression also reduced susceptibility to cisplatin-induced apoptosis (probably due to an increased BCL2: BAX ratio), accelerated epithelial-mesenchymal transition (EMT)-related cell mobility, and enhanced cancer stem cell (CSC) properties (including increases in both spheroid formation and in the aldehyde dehydrogenase 1 (ALDH1)high population). In contrast, S100A4 KO generally induced the opposite phenotypes, although it did not affect migration capability. In clinical OCCC samples, high S100A4 expression was associated with a low frequency of cleaved poly-(ADP-ribose) polymerase 1-positive apoptotic cells, a reduced proliferative rate, and expression of high ALDH1 and vimentin levels. In addition, a high S100A4 score was a significant (but not independent) prognostic factor in OCCC. CONCLUSIONS Our findings suggest that S100A4 may be an unfavorable prognostic factor in OCCC, as it accelerates tumor progression and promotes chemoresistance through the modulation of proliferation, susceptibility to apoptosis, and EMT/CSC properties.
Collapse
Affiliation(s)
| | | | | | | | | | - Makoto Saegusa
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Japan; (M.H.); (A.Y.); (M.N.); (M.H.); (Y.O.)
| |
Collapse
|
3
|
Giroud C, Szommer T, Coxon C, Monteiro O, Grimes T, Zarganes-Tzitzikas T, Christott T, Bennett J, Buchan K, Brennan PE, Fedorov O. Covalent Inhibitors of S100A4 Block the Formation of a Pro-Metastasis Non-Muscle Myosin 2A Complex. J Med Chem 2024; 67:18943-18956. [PMID: 39425667 PMCID: PMC11571109 DOI: 10.1021/acs.jmedchem.4c01320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
The S100 protein family functions as protein-protein interaction adaptors regulated by Ca2+ binding. Formation of various S100 complexes plays a central role in cell functions, from calcium homeostasis to cell signaling, and is implicated in cell growth, migration, and tumorigenesis. We established a suite of biochemical and cellular assays for small molecule screening based on known S100 protein-protein interactions. From 25 human S100 proteins, we focused our attention on S100A4 because of its well-established role in cancer progression and metastasizes by interacting with nonmuscle myosin II (NMII). We identified several potent and selective inhibitors of this interaction and established the covalent nature of binding, confirmed by mass spectrometry and crystal structures. 5b showed on-target activity in cells and inhibition of cancer cell migration. The identified S100A4 inhibitors can serve as a basis for the discovery of new cancer drugs operating via a novel mode of action.
Collapse
Affiliation(s)
- Charline Giroud
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Tamas Szommer
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Carmen Coxon
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Octovia Monteiro
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Thomas Grimes
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
- Alzheimer’s
Research UK Oxford Drug Discovery Institute, NDM Research Building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Tryfon Zarganes-Tzitzikas
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
- Alzheimer’s
Research UK Oxford Drug Discovery Institute, NDM Research Building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Thomas Christott
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - James Bennett
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Karly Buchan
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Paul E. Brennan
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Oleg Fedorov
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| |
Collapse
|
4
|
Quiralte M, Barquín A, Yagüe-Fernández M, Navarro P, Grazioso TP, Sevillano-Fernández E, Rodriguez-Moreno JF, Balarezo-Saldivar A, Peinado H, Izquierdo E, Millán C, López-Carrasco I, Prieto M, Madurga R, Fernández-Miranda I, Ruiz-Llorente S, García-Donas J. Proteomic profiles of peritoneal fluid-derived small extracellular vesicles correlate with patient outcome in ovarian cancer. J Clin Invest 2024; 134:e176161. [PMID: 38564289 PMCID: PMC11093605 DOI: 10.1172/jci176161] [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: 10/04/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer-derived small extracellular vesicles (sEVs) are capable of modifying the tumor microenvironment and promoting tumor progression. Ovarian cancer (OvCa) is a lethal malignancy that preferentially spreads through the abdominal cavity. Thus, the secretion of such vesicles into the peritoneal fluid could be a determinant factor in the dissemination and behavior of this disease. We designed a prospective observational study to assess the impact of peritoneal fluid-derived sEVs (PFD-sEVs) in OvCa clinical outcome. For this purpose, 2 patient cohorts were enrolled: patients with OvCa who underwent a diagnostic or cytoreductive surgery and nononcological patients, who underwent abdominal surgery for benign gynecological conditions and acted as the control group. Systematic extraction of PFD-sEVs from surgical samples enabled us to observe significant quantitative and qualitative differences associated with cancer diagnosis, disease stage, and platinum chemosensitivity. Proteomic profiling of PFD-sEVs led to the identification of molecular pathways and proteins of interest and to the biological validation of S100A4 and STX5. In addition, unsupervised analysis of PFD-sEV proteomic profiles in high-grade serous ovarian carcinomas (HGSOCs) revealed 2 clusters with different outcomes in terms of overall survival. In conclusion, comprehensive characterization of PFD-sEV content provided a prognostic value with potential implications in HGSOC clinical management.
Collapse
Affiliation(s)
- Miguel Quiralte
- Laboratory of Innovation in Oncology, Clara Campal Comprehensive Cancer Centre (HM CIOCC), HM Sanchinarro University Hospital, Madrid, Spain
- Institute of Applied Molecular Medicine, Faculty of Medicine, Universidad San Pablo–CEU, Alcorcón, Madrid, Spain
| | - Arantzazu Barquín
- Laboratory of Innovation in Oncology, Clara Campal Comprehensive Cancer Centre (HM CIOCC), HM Sanchinarro University Hospital, Madrid, Spain
- HM CIOCC, HM Sanchinarro University Hospital, Madrid, Spain
| | - Mónica Yagüe-Fernández
- Laboratory of Innovation in Oncology, Clara Campal Comprehensive Cancer Centre (HM CIOCC), HM Sanchinarro University Hospital, Madrid, Spain
| | - Paloma Navarro
- Laboratory of Innovation in Oncology, Clara Campal Comprehensive Cancer Centre (HM CIOCC), HM Sanchinarro University Hospital, Madrid, Spain
- Institute of Applied Molecular Medicine, Faculty of Medicine, Universidad San Pablo–CEU, Alcorcón, Madrid, Spain
| | - Tatiana P. Grazioso
- Laboratory of Innovation in Oncology, Clara Campal Comprehensive Cancer Centre (HM CIOCC), HM Sanchinarro University Hospital, Madrid, Spain
| | - Elena Sevillano-Fernández
- Laboratory of Innovation in Oncology, Clara Campal Comprehensive Cancer Centre (HM CIOCC), HM Sanchinarro University Hospital, Madrid, Spain
- HM CIOCC, HM Sanchinarro University Hospital, Madrid, Spain
| | - Juan F. Rodriguez-Moreno
- Laboratory of Innovation in Oncology, Clara Campal Comprehensive Cancer Centre (HM CIOCC), HM Sanchinarro University Hospital, Madrid, Spain
- HM CIOCC, HM Sanchinarro University Hospital, Madrid, Spain
| | - Alejandra Balarezo-Saldivar
- Laboratory of Innovation in Oncology, Clara Campal Comprehensive Cancer Centre (HM CIOCC), HM Sanchinarro University Hospital, Madrid, Spain
- Institute of Applied Molecular Medicine, Faculty of Medicine, Universidad San Pablo–CEU, Alcorcón, Madrid, Spain
| | - Héctor Peinado
- Microenvironment and Metastasis Laboratory, Molecular Oncology Program, Spanish National Cancer Research Centre, Madrid, Spain
| | - Elena Izquierdo
- Institute of Applied Molecular Medicine, Faculty of Medicine, Universidad San Pablo–CEU, Alcorcón, Madrid, Spain
| | - Carlos Millán
- Gynecologic Unit, HM Montepríncipe University Hospital, Boadilla del Monte, Madrid, Spain
| | - Irene López-Carrasco
- Gynecologic Unit, HM Montepríncipe University Hospital, Boadilla del Monte, Madrid, Spain
| | - Mario Prieto
- Department of Pathological Anatomy, Therapeutic Targets Laboratory, HM Sanchinarro University Hospital, Madrid, Spain
| | - Rodrigo Madurga
- Faculty of Experimental Sciences, Francisco de Vitoria University, Pozuelo de Alarcón, Madrid, Spain
| | - Ismael Fernández-Miranda
- R&D Oncology Business Unit, Pharmacogenomic and Cell Biology Departments, PharmaMar, Colmenar Viejo, Madrid, Spain
| | - Sergio Ruiz-Llorente
- Laboratory of Innovation in Oncology, Clara Campal Comprehensive Cancer Centre (HM CIOCC), HM Sanchinarro University Hospital, Madrid, Spain
- Department of Biomedicine and Biotechnology, Genetics Area, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - Jesús García-Donas
- Laboratory of Innovation in Oncology, Clara Campal Comprehensive Cancer Centre (HM CIOCC), HM Sanchinarro University Hospital, Madrid, Spain
- Institute of Applied Molecular Medicine, Faculty of Medicine, Universidad San Pablo–CEU, Alcorcón, Madrid, Spain
- HM CIOCC, HM Sanchinarro University Hospital, Madrid, Spain
| |
Collapse
|
5
|
Nakagawa M, Matsumoto T, Yokoi A, Hashimura M, Oguri Y, Konno R, Ishibashi Y, Ito T, Ohhigata K, Harada Y, Fukagawa N, Kodera Y, Saegusa M. Interaction between membranous EBP50 and myosin 9 as a favorable prognostic factor in ovarian clear cell carcinoma. Mol Oncol 2023; 17:2168-2182. [PMID: 37539980 PMCID: PMC10552901 DOI: 10.1002/1878-0261.13503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/29/2023] [Accepted: 08/01/2023] [Indexed: 08/05/2023] Open
Abstract
Ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50) is a scaffold protein that is required for epithelial polarity. Knockout (KO) of membranous EBP50 (Me-EBP50) in ovarian clear cell carcinoma (OCCC) cells induced an epithelial-mesenchymal transition (EMT)-like phenotype, along with decreased proliferation, accelerated migration capability, and induction of cancer stem cell (CSC)-like properties. Shotgun proteomics analysis of proteins that co-immunoprecipitated with EBP50 revealed that Me-EBP50 strongly interacts with myosin 9 (MYH9). Specific inhibition of MYH9 with blebbistatin phenocopied Me-EBP50 KO, and blebbistatin treatment potentiated the effects of Me-EBP50 KO. In OCCC cells from clinical samples, Me-EBP50 and MYH9 were co-localized at the apical plasma membrane. Patients with a combination of Me-EBP50-high and MYH9-high scores had the best prognosis for overall and progression-free survival. Our data suggest that Me-EBP50 has tumor-suppressive effects through the establishment and maintenance of epithelial polarization. By contrast, loss of Me-EBP50 expression induces EMT-like phenotypes, probably due to MYH9 dysfunction; this results in increased cell mobility and enhanced CSC-like properties, which in turn promote OCCC progression.
Collapse
Affiliation(s)
- Mayu Nakagawa
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Toshihide Matsumoto
- Department of PathologyKitasato University School of Allied Health ScienceSagamiharaJapan
| | - Ako Yokoi
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Miki Hashimura
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Yasuko Oguri
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Ryo Konno
- Center for Disease Proteomics, School of ScienceKitasato UniversitySagamiharaJapan
| | - Yu Ishibashi
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Takashi Ito
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Kensuke Ohhigata
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Yohei Harada
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Naomi Fukagawa
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Yoshio Kodera
- Center for Disease Proteomics, School of ScienceKitasato UniversitySagamiharaJapan
| | - Makoto Saegusa
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| |
Collapse
|
6
|
Huang C, Zheng D, Fu C, Cai Z, Zhang H, Xie Z, Luo L, Li H, Huang Y, Chen J. Secreted S100A4 causes asthmatic airway epithelial barrier dysfunction induced by house dust mite extracts via activating VEGFA/VEGFR2 pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:1431-1444. [PMID: 36883729 DOI: 10.1002/tox.23776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 02/21/2023] [Accepted: 02/25/2023] [Indexed: 05/18/2023]
Abstract
The airway epithelial barrier dysfunction plays a crucial role in pathogenesis of asthma and causes the amplification of downstream inflammatory signal pathway. S100 calcium binding protein A4 (S100A4), which promotes metastasis, have recently been discovered as an effective inflammatory factor and elevated in bronchoalveolar lavage fluid in asthmatic mice. Vascular endothelial growth factor-A (VEGFA), is considered as vital regulator in vascular physiological activities. Here, we explored the probably function of S100A4 and VEGFA in asthma model dealt with house dust mite (HDM) extracts. Our results showed that secreted S100A4 caused epithelial barrier dysfunction, airway inflammation and the release of T-helper 2 cytokines through the activation of VEGFA/VEGFR2 signaling pathway, which could be partial reversed by S100A4 polyclonal antibody, niclosamide and S100A4 knockdown, representing a potential therapeutic target for airway epithelial barrier dysfunction in asthma.
Collapse
Affiliation(s)
- Chaowen Huang
- Department of Pulmonary and Critical Care Medicine, Jiangmen Institute of Respiratory Disease, Jiangmen Central Hospital, Jiangmen, China
| | - Dongyan Zheng
- Department of Environmental and Occupational Health, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Chunlai Fu
- Department of Emergency Intensive Care Unit, Affiliated Dongguan People's Hospital, Southern Medical University, Dongguan, China
| | - Ziwei Cai
- Department of Environmental and Occupational Health, School of Public Health, Guangdong Medical University, Dongguan, China
| | - He Zhang
- Department of Environmental and Occupational Health, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Zhefan Xie
- Department of Emergency Intensive Care Unit, Affiliated Dongguan People's Hospital, Southern Medical University, Dongguan, China
| | - Lishan Luo
- Department of Respiratory and Critical Care Medicine, Huizhou Municipal Central Hospital, Huizhou, China
| | - Huifang Li
- Department of Environmental and Occupational Health, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Yanming Huang
- Department of Pulmonary and Critical Care Medicine, Jiangmen Institute of Respiratory Disease, Jiangmen Central Hospital, Jiangmen, China
| | - Jialong Chen
- Department of Environmental and Occupational Health, School of Public Health, Guangdong Medical University, Dongguan, China
| |
Collapse
|
7
|
Inoue A, Matsumoto T, Ito Y, Saegusa M, Takahashi H. TP53 positivity combined with high fibrinogen expression defines a subtype of oral squamous cell carcinoma with an unfavorable prognosis. Hum Pathol 2022; 130:25-35. [PMID: 36441045 DOI: 10.1016/j.humpath.2022.10.008] [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: 06/30/2022] [Revised: 10/04/2022] [Accepted: 10/20/2022] [Indexed: 11/04/2022]
Abstract
The number of deaths due to oral squamous cell carcinoma (OSCC), a malignant tumor of the oral cavity, is on the increase. We examined fibrinogen (FIB) expression in patients with OSCC and developed novel immunoprofile classification methods that include FIB. The plasma FIB level in patients with OSCC was elevated compared with that in patients with non-tumor oral disease (non-T); using a cut-off point of 342 mg/dL, we found the area under the curve-receiver operating characteristic level for OSCC was 0.745. Similarly, FIB expression in OSCC tissues was significantly higher compared with that in non-T tissues. Hierarchical clustering based on the immunoprofile of several markers including FIB, p53, and p16 revealed four groups that could be used to categorize OSCC cases (referred to as immunoprofile subtypes [IPS], I-IV). Tumors in IPS-II, which were FIB+/p53+, were associated with a significantly worse overall survival (OS) when compared with the other subtypes. We conclude that our IPS classification system can facilitate prognostic evaluation in OSCC, and that quantification of FIB is an important component of the classification strategy for this disease.
Collapse
Affiliation(s)
- Akemi Inoue
- Department of Pathology, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan
| | - Toshihide Matsumoto
- Department of Pathology, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan.
| | - Yuka Ito
- Department of Pathology, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan
| | - Makoto Saegusa
- Department of Pathology, School of Medicine, Kitasato University Sagamihara, Kanagawa 252-0374, Japan
| | - Hiroyuki Takahashi
- Department of Pathology, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa 252-0373, Japan
| |
Collapse
|
8
|
PTEN overexpression and nuclear β-catenin stabilization promote morular differentiation through induction of epithelial-mesenchymal transition and cancer stem cell-like properties in endometrial carcinoma. Cell Commun Signal 2022; 20:181. [PMID: 36411429 PMCID: PMC9677676 DOI: 10.1186/s12964-022-00999-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/23/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Although a lack of functional PTEN contributes to tumorigenesis in a wide spectrum of human malignancies, little is known about the functional role of its overexpression in the tumors. The current study focused on PTEN overexpression in endometrial carcinoma (Em Ca). METHODS The functional impact of PTEN overexpression was assessed by Em Ca cell lines. Immunohistochemical analyses were also conducted using 38 Em Ca with morular lesions. RESULTS Em Ca cell lines stably overexpressing PTEN (H6-PTEN) exhibited epithelial-mesenchymal transition (EMT)-like features, probably through β-catenin/Slug-meditated suppression of E-cadherin. PTEN overexpression also inhibited cell proliferation, accelerated cellular senescence, increased apoptotic features, and enhanced migration capability. Moreover, H6-PTEN cells exhibited cancer stem cell (CSC)-like properties, along with high expression of aldehyde dehydrogenase 1 and CD44s, a large ALDH 1high population, enriched spheroid formation, and β-catenin-mediated upregulation of cyclin D2, which is required for persistent CSC growth. In clinical samples, immunoreactivities for PTEN, as well as CSC-related molecules, were significantly higher in morular lesions as compared to the surrounding carcinomas. PTEN score was positively correlated with expression of nuclear β-catenin, cytoplasmic CD133, and CD44v6, and negatively with cell proliferation. Finally, estrogen receptor-α (ERα)-dependent expression of Ezrin-radixin-moesin-binding phophoprotein-50 (EBP50), a multifunctional scaffolding protein, acts as a negative regulator of morular formation by Em Ca cells through interacting with PTEN and β-catenin. CONCLUSION In the abscess of ERα/EBP50 expression, PTEN overexpression and nuclear β-catenin stabilization promote the establishment and maintenance of morular phenotype associated with EMT/CSC-like features in Em Ca cells. Video Abstract.
Collapse
|
9
|
A functional role of S100A4/non-muscle myosin IIA axis for pro-tumorigenic vascular functions in glioblastoma. Cell Commun Signal 2022; 20:46. [PMID: 35392912 PMCID: PMC8991692 DOI: 10.1186/s12964-022-00848-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/16/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most aggressive form of brain tumor and has vascular-rich features. The S100A4/non-muscle myosin IIA (NMIIA) axis contributes to aggressive phenotypes in a variety of human malignancies, but little is known about its involvement in GBM tumorigenesis. Herein, we examined the role of the S100A4/NMIIA axis during tumor progression and vasculogenesis in GBM. METHODS We performed immunohistochemistry for S100A4, NMIIA, and two hypoxic markers, hypoxia-inducible factor-1α (HIF-1α) and carbonic anhydrase 9 (CA9), in samples from 94 GBM cases. The functional impact of S100A4 knockdown and hypoxia were also assessed using a GBM cell line. RESULTS In clinical GBM samples, overexpression of S100A4 and NMIIA was observed in both non-pseudopalisading (Ps) and Ps (-associated) perinecrotic lesions, consistent with stabilization of HIF-1α and CA9. CD34(+) microvascular densities (MVDs) and the interaction of S100A4 and NMIIA were significantly higher in non-Ps perinecrotic lesions compared to those in Ps perinecrotic areas. In non-Ps perinecrotic lesions, S100A4(+)/HIF-1α(-) GBM cells were recruited to the surface of preexisting host vessels in the vascular-rich areas. Elevated vascular endothelial growth factor A (VEGFA) mRNA expression was found in S100A4(+)/HIF-1α(+) GBM cells adjacent to the vascular-rich areas. In addition, GBM patients with high S100A4 protein expression had significantly worse OS and PFS than did patients with low S100A4 expression. Knockdown of S100A4 in the GBM cell line KS-1 decreased migration capability, concomitant with decreased Slug expression; the opposite effects were elicited by blebbistatin-dependent inhibition of NMIIA. CONCLUSION S100A4(+)/HIF-1α(-) GBM cells are recruited to (and migrate along) preexisting vessels through inhibition of NMIIA activity. This is likely stimulated by extracellular VEGF that is released by S100A4(+)/HIF-1α(+) tumor cells in non-Ps perinecrotic lesions. In turn, these events engender tumor progression via acceleration of pro-tumorigenic vascular functions. Video abstract.
Collapse
|
10
|
Nakagawa M, Higuchi S, Hashimura M, Oguri Y, Matsumoto T, Yokoi A, Ishibashi Y, Ito T, Saegusa M. Functional interaction between S100A1 and MDM2 may modulate p53 signaling in normal and malignant endometrial cells. BMC Cancer 2022; 22:184. [PMID: 35177036 PMCID: PMC8855586 DOI: 10.1186/s12885-022-09249-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/13/2022] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND S100A1 expression is deregulated in a variety of human malignancies, but its role in normal and malignant endometrial cells is unclear. METHODS We used endometrial carcinoma (Em Ca) cell lines to evaluate the physical and functional interaction of S100A1 with p53 and its negative regulator, mouse double minute 2 (MDM2). We also evaluated the expression of S100A1, p53, and MDM2 in clinical samples consisting of 89 normal endometrial and 189 Em Ca tissues. RESULTS S100A1 interacted with MDM2 but not p53 in Em Ca cell lines. Treatment of cells stably overexpressing S100A1 with Nutlin-3A, an inhibitor of the p53/MDM2 interaction, increased expression of p53-target genes including p21waf1 and BAX. S100A1 overexpression enhanced cellular migration, but also sensitized cells to the antiproliferative and proapoptotic effects of Adriamycin, a genotoxic agent; these phenotypes were abrogated when S100A1 was knocked down using shRNA. In clinical samples from normal endometrium, S100A1 expression was significantly higher in endometrial glandular cells of the middle/late secretory and menstrual stages when compared to cells in the proliferative phases; high S100A1 was also positively correlated with expression of MDM2 and p21waf1 and apoptotic status, and inversely correlated with Ki-67 scores. However, such correlations were absent in Em Ca tissues. CONCLUSION The interaction between S100A1 and MDM2 may modulate proliferation, susceptibility to apoptosis, and migration through alterations in p53 signaling in normal- but not malignant-endometrial cells.
Collapse
Affiliation(s)
- Mayu Nakagawa
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Shyoma Higuchi
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Miki Hashimura
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yasuko Oguri
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Toshihide Matsumoto
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Ako Yokoi
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yu Ishibashi
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Takashi Ito
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Makoto Saegusa
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.
| |
Collapse
|
11
|
Anaplastic Lymphoma Kinase Overexpression Is Associated with Aggressive Phenotypic Characteristics of Ovarian High-Grade Serous Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1837-1850. [PMID: 34214505 DOI: 10.1016/j.ajpath.2021.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022]
Abstract
Deregulated full-length anaplastic lymphoma kinase (ALK) overexpression has been found in some primary solid tumors, but little is known about its role in ovarian high-grade serous carcinoma (HGSC). Herein, we focused on the functional roles of ALK in HGSC. Cytoplasmic ALK immunoreactivity without chromosomal rearrangement and gene mutations was significantly higher in HGSC compared with non-HGSC type ovarian carcinomas, and was significantly associated with several unfavorable clinicopathologic factors and poor prognosis. HGSC cell lines stably overexpressing ALK exhibited increased cell proliferation, enhanced cancer stem cell features, and accelerated cell mobility, whereas these phenotypes were abrogated in ALK-knockdown cells. Expression of the nervous system-associated gene, ELAVL3, and the corresponding protein (commonly known as HuC) was significantly increased in cells overexpressing ALK. There was increased expression of Sox2 and Sox3 (genes associated with the neural progenitor population) in ALK-overexpressing but not ALK-knockdown cells. Furthermore, overexpression of Sox2 or Sox3 enhanced both ALK and ELAVL3 promoter activities, suggesting the existence of ALK/Sox/HuC signaling loops. Finally, ALK overexpression was due to increased expression of neuroendocrine markers, including synaptophysin, CD56, and BCL2, in HGSC tissues. These findings suggest that overexpression of full-length ALK may influence the biological behavior of HGSC through cooperation with ELAVL3 and Sox factors, leading to establishment and maintenance of the aggressive phenotypic characteristics of HGSC.
Collapse
|
12
|
Santolla MF, Talia M, Maggiolini M. S100A4 Is Involved in Stimulatory Effects Elicited by the FGF2/FGFR1 Signaling Pathway in Triple-Negative Breast Cancer (TNBC) Cells. Int J Mol Sci 2021; 22:ijms22094720. [PMID: 33946884 PMCID: PMC8124532 DOI: 10.3390/ijms22094720] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast tumor subtype characterized by poor clinical outcome. In recent years, numerous advancements have been made to better understand the biological landscape of TNBC, though appropriate targets still remain to be determined. In the present study, we have determined that the expression levels of FGF2 and S100A4 are higher in TNBC with respect to non-TNBC patients when analyzing “The Invasive Breast Cancer Cohort of The Cancer Genome Atlas” (TCGA) dataset. In addition, we have found that the gene expression of FGF2 is positively correlated with S100A4 in TNBC samples. Performing quantitative PCR, Western blot, CRISPR/Cas9 genome editing, promoter studies, immunofluorescence analysis, subcellular fractionation studies, and ChIP assays, we have also demonstrated that FGF2 induces in TNBC cells the upregulation and secretion of S100A4 via FGFR1, along with the ERK1/2–AKT–c-Rel transduction signaling. Using conditioned medium from TNBC cells stimulated with FGF2, we have also ascertained that the paracrine activation of the S100A4/RAGE pathway triggers angiogenic effects in vascular endothelial cells (HUVECs) and promotes the migration of cancer-associated fibroblasts (CAFs). Collectively, our data provide novel insights into the action of the FGF2/FGFR1 axis through S100A4 toward stimulatory effects elicited in TNBC cells.
Collapse
MESH Headings
- Antigens, Neoplasm/physiology
- Cell Movement/drug effects
- Culture Media, Conditioned/pharmacology
- Female
- Fibroblast Growth Factor 2/pharmacology
- Fibroblast Growth Factor 2/physiology
- Fibroblasts/pathology
- Gene Expression Regulation, Neoplastic/physiology
- Human Umbilical Vein Endothelial Cells
- Humans
- Mitogen-Activated Protein Kinases/physiology
- Neoplasm Proteins/physiology
- Neovascularization, Pathologic/physiopathology
- Paracrine Communication
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-rel/physiology
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/physiology
- S100 Calcium-Binding Protein A4/physiology
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Triple Negative Breast Neoplasms/blood supply
- Triple Negative Breast Neoplasms/physiopathology
- Tumor Cells, Cultured
Collapse
|