1
|
Wang L, Liu X, Lin Z, Xiao Z, Ming J. Clinical characteristics and prognostic factors analysis of patients stricken with double primary breast and ovarian cancer based on the SEER database. Gland Surg 2025; 14:405-420. [PMID: 40256485 PMCID: PMC12004301 DOI: 10.21037/gs-24-480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/04/2025] [Indexed: 04/22/2025]
Abstract
Background Dual primary breast cancer (BC) and ovarian cancer (OC) represent a distinct subset of patients with diverse survival situation compared to those with a single primary BC or OC. Nonetheless, comprehensive research on their clinical characteristics and prognosis is lacking. This study conducted a retrospective analysis of clinical characteristics, survival outcomes, and prognostic factors of dual primary BC and OC patients. Methods We applied the National Cancer Institute Surveillance, Epidemiology, and End Results (SEER) database to identify patients with dual primary BC and primary OC (DPBOC) from 2000 to 2019, and divided patients into two groups: the BC-first group (BO group) and the OC-first group (OB group). Moreover, we employed Kaplan-Meier method to assess overall survival (OS), breast cancer-specific survival (BCSS), and ovarian cancer-specific survival (OCSS), and the Cox proportional hazards model to analyze prognostic factors. Results There were 1,074 patients enrolled, 665 in the BO group and 409 in the OB group. The median time interval was for 48 (range, 0-228) months. There were significant differences in serous carcinoma and OC tumor stage between the two groups (P<0.001; P<0.001). There was no significant difference in BCSS between the two groups (Log-rank P=0.67), but the BO group had inferior OS and OCSS than the OB group (Log-rank P<0.001). Patients with an interval of ≥48 months had a significantly lower risk of death [hazard ratio (HR) =0.323, 95% confidence interval (CI): 0.264-0.395, P<0.001; HR =0.527, 95% CI: 0.305-0.908, P=0.02; HR =0.709, 95% CI: 0.560-0.897, P=0.004]. Conclusions OC primarily determines the survival outcomes of DPBOC. Patients with BC as the first primary cancer (FPC) have a worse prognosis than patients with OC as FPC. After a diagnosis of BC or OC, we should pay close attention to another site, particularly after BC diagnosis, and monitor screening for ovarian lesions as early as feasible, as well as strengthening the treatment for OC.
Collapse
Affiliation(s)
- Lu Wang
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaowei Liu
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zijing Lin
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhesi Xiao
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jia Ming
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Cao Z, Quazi S, Arora S, Osellame LD, Burvenich IJ, Janes PW, Scott AM. Cancer-associated fibroblasts as therapeutic targets for cancer: advances, challenges, and future prospects. J Biomed Sci 2025; 32:7. [PMID: 39780187 PMCID: PMC11715488 DOI: 10.1186/s12929-024-01099-2] [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: 04/05/2024] [Accepted: 11/09/2024] [Indexed: 01/11/2025] Open
Abstract
Research into cancer treatment has been mainly focused on developing therapies to directly target cancer cells. Over the past decade, extensive studies have revealed critical roles of the tumour microenvironment (TME) in cancer initiation, progression, and drug resistance. Notably, cancer-associated fibroblasts (CAFs) have emerged as one of the primary contributors in shaping TME, creating a favourable environment for cancer development. Many preclinical studies have identified promising targets on CAFs, demonstrating remarkable efficacy of some CAF-targeted treatments in preclinical models. Encouraged by these compelling findings, therapeutic strategies have now advanced into clinical evaluation. We aim to provide a comprehensive review of relevant subjects on CAFs, including CAF-related markers and targets, their multifaceted roles, and current landscape of ongoing clinical trials. This knowledge can guide future research on CAFs and advocate for clinical investigations targeting CAFs.
Collapse
Affiliation(s)
- Zhipeng Cao
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia.
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia.
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, 3084, Australia.
| | - Sadia Quazi
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Sakshi Arora
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Laura D Osellame
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Ingrid J Burvenich
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Peter W Janes
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, 3084, Australia.
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia.
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, 3084, Australia.
- Department of Medicine, University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
3
|
Yu S, Wang S, Wang X, Xu X. The axis of tumor-associated macrophages, extracellular matrix proteins, and cancer-associated fibroblasts in oncogenesis. Cancer Cell Int 2024; 24:335. [PMID: 39375726 PMCID: PMC11459962 DOI: 10.1186/s12935-024-03518-8] [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: 02/22/2024] [Accepted: 09/29/2024] [Indexed: 10/09/2024] Open
Abstract
The extracellular matrix (ECM) is a complex, dynamic network of multiple macromolecules that serve as a crucial structural and physical scaffold for neighboring cells. In the tumor microenvironment (TME), ECM proteins play a significant role in mediating cellular communication between cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs). Revealing the ECM modification of the TME necessitates the intricate signaling cascades that transpire among diverse cell populations and ECM proteins. The advent of single-cell sequencing has enabled the identification and refinement of specific cellular subpopulations, which has substantially enhanced our comprehension of the intricate milieu and given us a high-resolution perspective on the diversity of ECM proteins. However, it is essential to integrate single-cell data and establish a coherent framework. In this regard, we present a comprehensive review of the relationships among ECM, TAMs, and CAFs. This encompasses insights into the ECM proteins released by TAMs and CAFs, signaling integration in the TAM-ECM-CAF axis, and the potential applications and limitations of targeted therapies for CAFs. This review serves as a reliable resource for focused therapeutic strategies while highlighting the crucial role of ECM proteins as intermediates in the TME.
Collapse
Affiliation(s)
- Shuhong Yu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Siyu Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xuanyu Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ximing Xu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
4
|
Rozen EJ, Frantz W, Wigglesworth K, Vessella T, Zhou HS, Shohet JM. Blockade of Discoidin Domain Receptor Signaling with Sitravatinib Reveals DDR2 as a Mediator of Neuroblastoma Pathogenesis and Metastasis. Mol Cancer Ther 2024; 23:1124-1138. [PMID: 38670553 DOI: 10.1158/1535-7163.mct-23-0741] [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: 10/25/2023] [Revised: 02/06/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
Oncogene-driven expression and activation of receptor tyrosine kinases promotes tumorigenesis and contributes to drug resistance. Increased expression of the kinases discoidin domain receptor 2 (DDR2), RET Proto-Oncogene (RET), Platelet Derived Growth Factor Receptor Alpha (PDGFRA), KIT Proto-Oncogene (KIT), MET Proto-Oncogene (MET), and anaplastic lymphoma kinase (ALK) independently correlate with decreased overall survival and event free survival of pediatric neuroblastoma. The multikinase inhibitor sitravatinib targets DDR2, RET, PDGFRA, KIT, and MET with low nanomolar activity and we therefore tested its efficacy against orthotopic and syngeneic tumor models. Sitravatinib markedly reduced cell proliferation and migration in vitro independently of N-Myc proto-oncogene (MYCN), ALK, or c-Myc proto-oncogene status and inhibited proliferation and metastasis of human orthotopic xenografts. Oral administration of sitravatinib to homozygous Th-MYCN transgenic mice (Th-MYCN+/+) after tumor initiation completely arrested further tumor development with no mice dying of disease while maintained on sitravatinib treatment (control cohort 57 days median time to sacrifice). Among these top kinases, DDR2 expression has the strongest correlation with poor survival and high stage at diagnosis and the highest sensitivity to the drug. We confirmed on-target inhibition of collagen-mediated activation of DDR2. Genetic knockdown of DDR2 partially phenocopies sitravatinib treatment, limiting tumor development and metastasis across tumor models. Analysis of single-cell sequencing data demonstrated that DDR2 is restricted to mesenchymal-type tumor subpopulations and is enriched in Schwann cell precursor subpopulations found in high-risk disease. These data define an unsuspected role for sitravatinib as a therapeutic agent in neuroblastoma and reveal a novel function for DDR2 as a driver of tumor growth and metastasis.
Collapse
Affiliation(s)
- Esteban J Rozen
- Crnic Institute Boulder Branch, BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado
| | - William Frantz
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Kim Wigglesworth
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Theadora Vessella
- Department of Chemical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Hong S Zhou
- Department of Chemical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Jason M Shohet
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
5
|
Chen J, Yang L, Ma Y, Zhang Y. Recent advances in understanding the immune microenvironment in ovarian cancer. Front Immunol 2024; 15:1412328. [PMID: 38903506 PMCID: PMC11188340 DOI: 10.3389/fimmu.2024.1412328] [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: 04/04/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024] Open
Abstract
The occurrence of ovarian cancer (OC) is a major factor in women's mortality rates. Despite progress in medical treatments, like new drugs targeting homologous recombination deficiency, survival rates for OC patients are still not ideal. The tumor microenvironment (TME) includes cancer cells, fibroblasts linked to cancer (CAFs), immune-inflammatory cells, and the substances these cells secrete, along with non-cellular components in the extracellular matrix (ECM). First, the TME mainly plays a role in inhibiting tumor growth and protecting normal cell survival. As tumors progress, the TME gradually becomes a place to promote tumor cell progression. Immune cells in the TME have attracted much attention as targets for immunotherapy. Immune checkpoint inhibitor (ICI) therapy has the potential to regulate the TME, suppressing factors that facilitate tumor advancement, reactivating immune cells, managing tumor growth, and extending the survival of patients with advanced cancer. This review presents an outline of current studies on the distinct cellular elements within the OC TME, detailing their main functions and possible signaling pathways. Additionally, we examine immunotherapy rechallenge in OC, with a specific emphasis on the biological reasons behind resistance to ICIs.
Collapse
Affiliation(s)
- Jinxin Chen
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Lu Yang
- Department of Internal Medicine, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yiming Ma
- Department of Medical Oncology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
- Liaoning Key Laboratory of Gastrointestinal Cancer Translational Research, Shenyang, Liaoning, China
| | - Ye Zhang
- Department of Radiation Oncology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
6
|
Han J, Lyu L. Identification of the biological functions and chemo-therapeutic responses of ITGB superfamily in ovarian cancer. Discov Oncol 2024; 15:198. [PMID: 38814534 PMCID: PMC11139846 DOI: 10.1007/s12672-024-01047-4] [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: 03/31/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Patients with ovarian cancer (OC) tend to face a poor prognosis due to a lack of typical symptoms and a high rate of recurrence and chemo-resistance. Therefore, identifying representative and reliable biomarkers for early diagnosis and prediction of chemo-therapeutic responses is vital for improving the prognosis of OC. METHODS Expression levels, IHC staining, and subcellular distribution of eight ITGBs were analyzed using The Cancer Genome Atlas (TCGA)-Ovarian Serous Cystadenocarcinoma (OV) database, GEO DataSets, and the HPA website. PrognoScan and Univariate Cox were used for prognostic analysis. TIDE database, TIMER database, and GSCA database were used to analyze the correlation between immune functions and ITGBs. Consensus clustering analysis was performed to subtype OC patients in the TCGA database. LASSO regression was used to construct the predictive model. The Cytoscape software was used for identifying hub genes. The 'pRRophetic' R package was applied to predict chemo-therapeutic responses of ITGBs. RESULTS ITGBs were upregulated in OC tissues except ITGB1 and ITGB3. High expression of ITGBs correlated with an unfavorable prognosis of OC except ITGB2. In OC, there was a strong correlation between immune responses and ITGB2, 6, and 7. In addition, the expression matrix of eight ITGBs divided the TCGA-OV database into two subgroups. Subgroup A showed upregulation of eight ITGBs. The predictive model distinguishes OC patients from favorable prognosis to poor prognosis. Chemo-therapeutic responses showed that ITGBs were able to predict responses of common chemo-therapeutic drugs for patients with OC. CONCLUSIONS This article provides evidence for predicting prognosis, immuno-, and chemo-therapeutic responses of ITGBs in OC and reveals related biological functions of ITGBs in OC.
Collapse
Affiliation(s)
- Jiawen Han
- Department of Nutrition, Jinshan Hospital, Fudan University, 1508 Longhang Road, Jinshan District, Shanghai, 201508, China
| | - Lin Lyu
- Department of Nutrition, Jinshan Hospital, Fudan University, 1508 Longhang Road, Jinshan District, Shanghai, 201508, China.
| |
Collapse
|
7
|
Shen L, Li A, Cui J, Liu H, Zhang S. Integration of single-cell RNA-seq and bulk RNA-seq data to construct and validate a cancer-associated fibroblast-related prognostic signature for patients with ovarian cancer. J Ovarian Res 2024; 17:82. [PMID: 38627854 PMCID: PMC11020192 DOI: 10.1186/s13048-024-01399-z] [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: 05/09/2023] [Accepted: 03/21/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND To establish a prognostic risk profile for ovarian cancer (OC) patients based on cancer-associated fibroblasts (CAFs) and gain a comprehensive understanding of their role in OC progression, prognosis, and therapeutic efficacy. METHODS Data on OC single-cell RNA sequencing (scRNA-seq) and total RNA-seq were collected from the GEO and TCGA databases. Seurat R program was used to analyze scRNA-seq data and identify CAFs clusters corresponding to CAFs markers. Differential expression analysis was performed on the TCGA dataset to identify prognostic genes. A CAF-associated risk signature was designed using Lasso regression and combined with clinicopathological variables to develop a nomogram. Functional enrichment and the immune landscape were also analyzed. RESULTS Five CAFs clusters were identified in OC using scRNA-seq data, and 2 were significantly associated with OC prognosis. Seven genes were selected to develop a CAF-based risk signature, primarily associated with 28 pathways. The signature was a key independent predictor of OC prognosis and relevant in predicting the results of immunotherapy interventions. A novel nomogram combining CAF-based risk and disease stage was developed to predict OC prognosis. CONCLUSION The study highlights the importance of CAFs in OC progression and suggests potential for innovative treatment strategies. A CAF-based risk signature provides a highly accurate prediction of the prognosis of OC patients, and the developed nomogram shows promising results in predicting the OC prognosis.
Collapse
Affiliation(s)
- Liang Shen
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, 67 Dongchang West Road, Liaocheng, Shandong, 252000, P.R. China
- Shandong University, Jinan, P.R. China
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, Shandong, 250021, P.R. China
| | - Aihua Li
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, 67 Dongchang West Road, Liaocheng, Shandong, 252000, P.R. China.
| | - Jing Cui
- Department of Oral and Maxillofacial Surgery, Jinan Stomatology Hospital, 101 Jingliu Road, Jinan, Shandong, 250001, P.R. China
- Central Laboratory of Jinan Stamotological Hospital, Jinan Key Laboratory of Oral Tissue Regeneration, 101 Jingliu Road, Jinan, Shandong, 250001, P.R. China
| | - Haixia Liu
- Department of Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, Shandong, 250021, P.R. China
| | - Shiqian Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, P.R. China.
| |
Collapse
|
8
|
Jia X, Li Z, Zhou R, Feng W, Yi L, Zhang H, Chen B, Li Q, Huang S, Zhu X. Single cell and bulk RNA sequencing identifies tumor microenvironment subtypes and chemoresistance-related IGF1 + cancer-associated fibroblast in gastric cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167123. [PMID: 38484940 DOI: 10.1016/j.bbadis.2024.167123] [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: 08/02/2023] [Revised: 03/03/2024] [Accepted: 03/11/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND The tumor microenvironment (TME) significantly influences prognosis and drug resistance in various tumors, yet its heterogeneity and the mechanisms affecting therapeutic response remain unclear in gastric cancer (GC). METHODS The heterogenous TME were explored with single-cell RNA-sequencing (scRNA-seq) data of 50 primary GC samples. We then identified four GC TME subtypes with nonnegative matrix factorization (NMF) and constructed a pearson nearest-centroid classifier based on subtype-specific upregulated genes. Genomic features and clinical significance of four subtypes were comprehensively evaluated. We reclustered fibroblasts to identify cancer-associated fibroblast (CAF) subtype associated with poor clinical outcomes. RT-qPCR and double immunofluorescence staining were applied to validate the findings. Cellchat analysis elucidated potential molecular mechanisms of the CAF subtype in GC disease progression and chemotherapy resistance. FINDINGS The GC TME exhibited high heterogeneity, influencing chemo-sensitivity. Four TME-based subtypes predicting response to immunotherapy and chemotherapy were identified and validated in 1406 GC patients. Among which, ISG1 subtype displayed higher fibroblasts infiltration and heightened oncogenic pathways, and inferior response to chemotherapy with unfavorable prognosis. Microsatellite instability-high (MSI-H) GCs within four TME subtypes showed immunological heterogeneity. We then reported an IGF1-overexpressing CAF was associated with chemo-resistance and GC recurrence. Cell communication analysis revealed IGF1+ CAF may induce drug-resistant phenotypes in tumor cells through IGF1-α6β4 integrin ligand-receptor binding and activation of EMT biological process. INTERPRETATION We identified four TME-based subtypes with different clinical outcomes and IGF1+ CAFs contributing to poor clinical outcomes in GC, which might provide guidance for individualized treatment and facilitate the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Xiya Jia
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ziteng Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Runye Zhou
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wanjing Feng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lixia Yi
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hena Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Bing Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Qin Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shenglin Huang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Xiaodong Zhu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
9
|
Su C, Mo J, Dong S, Liao Z, Zhang B, Zhu P. Integrinβ-1 in disorders and cancers: molecular mechanisms and therapeutic targets. Cell Commun Signal 2024; 22:71. [PMID: 38279122 PMCID: PMC10811905 DOI: 10.1186/s12964-023-01338-3] [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: 08/23/2023] [Accepted: 09/27/2023] [Indexed: 01/28/2024] Open
Abstract
Integrinβ-1 (ITGB1) is a crucial member of the transmembrane glycoprotein signaling receptor family and is also central to the integrin family. It forms heterodimers with other ligands, participates in intracellular signaling and controls a variety of cellular processes, such as angiogenesis and the growth of neurons; because of its role in bidirectional signaling regulation both inside and outside the membrane, ITGB1 must interact with a multitude of substances, so a variety of interfering factors can affect ITGB1 and lead to changes in its function. Over the past 20 years, many studies have confirmed a clear causal relationship between ITGB1 dysregulation and cancer development and progression in a wide range of benign diseases and solid tumor types, which may imply that ITGB1 is a prognostic biomarker and a therapeutic target for cancer treatment that warrants further investigation. This review summarizes the biological roles of ITGB1 in benign diseases and cancers, and compiles the current status of ITGB1 function and therapy in various aspects of tumorigenesis and progression. Finally, future research directions and application prospects of ITGB1 are suggested. Video Abstract.
Collapse
Affiliation(s)
- Chen Su
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China
| | - Jie Mo
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China
| | - Shuilin Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China
| | - Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, People's Republic of China.
| | - Peng Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
10
|
Trono P, Ottavi F, Rosano' L. Novel insights into the role of Discoidin domain receptor 2 (DDR2) in cancer progression: a new avenue of therapeutic intervention. Matrix Biol 2024; 125:31-39. [PMID: 38081526 DOI: 10.1016/j.matbio.2023.12.003] [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: 10/27/2023] [Revised: 11/22/2023] [Accepted: 12/08/2023] [Indexed: 02/12/2024]
Abstract
Discoidin domain receptors (DDRs), including DDR1 and DDR2, are a unique class of receptor tyrosine kinases (RTKs) activated by collagens at the cell-matrix boundary interface. The peculiar mode of activation makes DDRs as key cellular sensors of microenvironmental changes, with a critical role in all physiological and pathological processes governed by collagen remodeling. DDRs are widely expressed in fetal and adult tissues, and experimental and clinical evidence has shown that their expression is deregulated in cancer. Strong findings supporting the role of collagens in tumor progression and metastasis have led to renewed interest in DDRs. However, despite an increasing number of studies, DDR biology remains poorly understood, particularly the less studied DDR2, whose involvement in cancer progression mechanisms is undoubted. Thus, the understanding of a wider range of DDR2 functions and related molecular mechanisms is expected. To date, several lines of evidence support DDR2 as a promising target in cancer therapy. Its involvement in key functions in the tumor microenvironment makes DDR2 inhibition particularly attractive to achieve simultaneous targeting of tumor and stromal cells, and tumor regression, which is beneficial for improving the response to different types of anti-cancer therapies, including chemo- and immunotherapy. This review summarizes current research on DDR2, focusing on its role in cancer progression through its involvement in tumor and stromal cell functions, and discusses findings that support the rationale for future development of direct clinical strategies targeting DDR2.
Collapse
Affiliation(s)
- Paola Trono
- Institute of Biochemistry and Cell Biology (IBBC)-CNR, Via E. Ramarini, 32, Monterotondo Scalo 00015 Rome
| | - Flavia Ottavi
- Institute of Molecular Biology and Pathology (IBPM)-CNR, Via degli Apuli 4, Rome 00185, Italy
| | - Laura Rosano'
- Institute of Molecular Biology and Pathology (IBPM)-CNR, Via degli Apuli 4, Rome 00185, Italy.
| |
Collapse
|
11
|
Akinjiyan FA, Ibitoye Z, Zhao P, Shriver LP, Patti GJ, Longmore GD, Fuh KC. DDR2-regulated arginase activity in ovarian cancer-associated fibroblasts promotes collagen production and tumor progression. Oncogene 2024; 43:189-201. [PMID: 37996700 PMCID: PMC10786713 DOI: 10.1038/s41388-023-02884-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/21/2023] [Accepted: 10/30/2023] [Indexed: 11/25/2023]
Abstract
Ovarian cancer has poor survival outcomes particularly for advanced stage, metastatic disease. Metastasis is promoted by interactions of stromal cells, such as cancer-associated fibroblasts (CAFs) in the tumor microenvironment (TME), with tumor cells. CAFs play a key role in tumor progression by remodeling the TME and extracellular matrix (ECM) to result in a more permissive environment for tumor progression. It has been shown that fibroblasts, in particular myofibroblasts, utilize metabolism to support ECM remodeling. However, the intricate mechanisms by which CAFs support collagen production and tumor progression are poorly understood. In this study, we show that the fibrillar collagen receptor, Discoidin Domain Receptor 2 (DDR2), promotes collagen production in human and mouse omental CAFs through arginase activity. CAFs with high DDR2 or arginase promote tumor colonization in the omentum. In addition, DDR2-depleted CAFs had decreased ornithine levels leading to decreased collagen production and polyamine levels compared to WT control CAFs. Tumor cell invasion was decreased in the presence CAF conditioned media (CM) depleted of DDR2 or arginase-1, and this invasion defect was rescued in the presence of CM from DDR2-depleted CAFs that constitutively overexpressed arginase-1. Similarly, the addition of exogenous polyamines to CM from DDR2-depleted CAFs led to increased tumor cell invasion. We detected SNAI1 protein at the promoter region of the arginase-1 gene, and DDR2-depleted CAFs had decreased levels of SNAI1 protein at the arginase-1 promoter region. Furthermore, high stromal arginase-1 expression correlated with poor survival in ovarian cancer patients. These findings highlight how DDR2 regulates collagen production by CAFs in the tumor microenvironment by controlling the transcription of arginase-1, and CAFs are a major source of arginase activity and L-arginine metabolites in ovarian cancer models.
Collapse
Affiliation(s)
- Favour A Akinjiyan
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Center for Reproductive Health Sciences, Washington University, St Louis, MO, 63110, USA
- ICCE Institute, Washington University, St Louis, MO, 63110, USA
- Department of Medicine (Oncology), Washington University, St. Louis, MO, 63110, USA
| | - Zainab Ibitoye
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Center for Reproductive Health Sciences, Washington University, St Louis, MO, 63110, USA
- ICCE Institute, Washington University, St Louis, MO, 63110, USA
- Department of Medicine (Oncology), Washington University, St. Louis, MO, 63110, USA
| | - Peinan Zhao
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Leah P Shriver
- Department of Medicine (Oncology), Washington University, St. Louis, MO, 63110, USA
- Department of Chemistry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Center for Metabolomics and Isotope Tracing, Washington University, St. Louis, MO, 63130, USA
| | - Gary J Patti
- Department of Medicine (Oncology), Washington University, St. Louis, MO, 63110, USA
- Department of Chemistry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Center for Metabolomics and Isotope Tracing, Washington University, St. Louis, MO, 63130, USA
| | - Gregory D Longmore
- ICCE Institute, Washington University, St Louis, MO, 63110, USA
- Department of Medicine (Oncology), Washington University, St. Louis, MO, 63110, USA
| | - Katherine C Fuh
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Center for Reproductive Health Sciences, Washington University, St Louis, MO, 63110, USA.
- Department of Obstetrics and Gynecology & Reproductive Sciences, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
12
|
Zhi D, Zhou K, Liu S, Yu W, Dong M, Yan C. METTL3/YTHDF1 m 6A axis promotes tumorigenesis by enhancing DDR2 expression in ovarian cancer. Pathol Res Pract 2024; 253:155047. [PMID: 38154356 DOI: 10.1016/j.prp.2023.155047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/06/2023] [Accepted: 09/15/2023] [Indexed: 12/30/2023]
Abstract
Ovarian cancer has the highest mortality among all gynecological malignancies. Therefore, it is urgent to determine the molecular mechanism of ovarian cancer progression. As the most prevalent modification of messenger RNA (mRNA), N6-Methyladenosine (m6A) modification is recognized as a key regulatory role in the progression of various tumors. However, the specific role of m6A and its related regulatory pathways in ovarian cancer (OV) remains unclear. In this study, we demonstrated that the METTL3/YTHDF1 m6A axis plays an important role in the progression of ovarian cancer. Depletion of METTL3/YTHDF1 impaired cancer proliferation and metastasis in vitro and in vivo. Mechanistically, The METTL3/YTHDF1 m6A axis directly binds to the mRNA of DDR2, thereby promoting the expression levels of the tumor promoter DDR2 and thus contributing to the progression of ovarian cancer. Collectively, our findings on the METTL3/YTHDF1/DDR2 m6A axis provide the insight into the underlying mechanism of ovarian carcinogenesis and highlight potential therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Duo Zhi
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150040, China
| | - Kun Zhou
- Beidahuang Industry Group General Hospital, Department of Clinical Laboratory, No. 235, Hashuang Road, Nangang District, Harbin, Heilongjiang, China
| | - Shuang Liu
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150040, China
| | - Wen Yu
- Jiamusi Medical Insurance Bureau Hospital, China
| | - Mei Dong
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150040, China.
| | - Caichuan Yan
- Department of Cancer Molecular and Biology, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
13
|
Zhao Z, Mak TK, Shi Y, Li K, Huo M, Zhang C. Integrative analysis of cancer-associated fibroblast signature in gastric cancer. Heliyon 2023; 9:e19217. [PMID: 37809716 PMCID: PMC10558323 DOI: 10.1016/j.heliyon.2023.e19217] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 10/10/2023] Open
Abstract
Background CAFs regulate the signaling of GC cells by promoting their migration, invasion, and proliferation and the function of immune cells as well as their location and migration in the TME by remodeling the extracellular matrix (ECM). This study explored the understanding of the heterogeneity of CAFs in TME and laid the groundwork for GC biomarker and precision treatment development. Methods The scRNA-seq and bulk RNA-seq datasets were obtained from GEO and TCGA. The prognostic significance of various CAFs subtypes was investigated using ssGSEA combined with Kaplan-Meier analysis. POSTN expression in GC tissues and CAFs was detected using immunohistochemistry, immunofluorescence, and Western blotting. Differential expression analysis identified the differentially expressed genes (DEGs) between normal and tumor samples in TCGA-STAD. Pearson correlation analysis identified DEGs associated with adverse prognosis CAF subtype, and univariate Cox regression analysis determined prognostic genes associated with CAFs. LASSO regression analysis and Multivariate Cox regression were used to build a prognosis model for CAFs. Results We identified five CAFs subtypes in GC, with the CAF_0 subtype associated with poor prognosis. The abundance of CAF_0 correlated with T stage, clinical stage, histological type, and immune cell infiltration levels. Periostin (POSTN) exhibited increased expression in both GC tissues and CAFs and was linked to poor prognosis in GC patients. Through LASSO and multivariate Cox regression analysis, three genes (CXCR4, MATN3, and KIF24) were selected to create the CAFs-score. We developed a nomogram to facilitate the clinical application of the CAFs-score. Notably, the CAFs signature showed significant correlations with immune cells, stromal components, and immunological scores, suggesting its pivotal role in the tumor microenvironment (TME). Furthermore, CAFs-score demonstrated prognostic value in assessing immunotherapy outcomes, highlighting its potential as a valuable biomarker to guide therapeutic decisions. Conclusion CAF_0 subtype in TME is the cause of poor prognosis in GC patients. Furthermore, CAFs-score constructed from the CAF_0 subtype can be used to determine the clinical prognosis, immune infiltration, clinicopathological characteristics, and assessment of personalized treatment of GC patients.
Collapse
Affiliation(s)
- Zidan Zhao
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Tsz Kin Mak
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yuntao Shi
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Kuan Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Mingyu Huo
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
14
|
Ge C, Li Y, Wu F, Ma P, Franceschi RT. Synthetic peptides activating discoidin domain receptor 2 and collagen-binding integrins cooperate to stimulate osteoblast differentiation of skeletal progenitor cells. Acta Biomater 2023; 166:109-118. [PMID: 37245640 PMCID: PMC10617013 DOI: 10.1016/j.actbio.2023.05.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Skeletal progenitor: collagen interactions are critical for bone development and regeneration. Both collagen-binding integrins and discoidin domain receptors (DDR1 and DDR2) function as collagen receptors in bone. Each receptor is activated by a distinct collagen sequence; GFOGER for integrins and GVMGFO for DDRs. Specific triple helical peptides containing each of these binding domains were evaluated for ability to stimulate DDR2 and integrin signaling and osteoblast differentiation. GVMGFO peptide stimulated DDR2 Y740 phosphorylation and osteoblast differentiation as measured by induction of osteoblast marker mRNAs and mineralization without affecting integrin activity. In contrast, GFOGER peptide stimulated focal adhesion kinase (FAK) Y397 phosphorylation, an early measure of integrin activation, and to a lesser extent osteoblast differentiation without affecting DDR2-P. Significantly, the combination of both peptides cooperatively enhanced both DDR2 and FAK signaling and osteoblast differentiation, a response that was blocked in Ddr2-deficient cells. These studies suggest that the development of scaffolds containing DDR and integrin-activating peptides may provide a new route for promoting bone regeneration. STATEMENT OF SIGNIFICANCE: A method for stimulating osteoblast differentiation of skeletal progenitor cells is described that uses culture surfaces coated with a collagen-derived triple-helical peptide to selectively activate discoidin domain receptors. When this peptide is combined with an integrin-activating peptide, synergistic stimulation of differentiation is seen. This approach of combining collagen-derived peptides to stimulate the two main collagen receptors in bone (DDR2 and collagen-binding integrins) provides a route for developing a new class of tissue engineering scaffolds for bone regeneration.
Collapse
Affiliation(s)
- Chunxi Ge
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 N. University Avenue, Ann Arbor, MI 48109-1078, USA
| | - Yiming Li
- Department of Biologic and Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, 1011 N. University Avenue, Ann Arbor, MI 48109-1078, USA
| | - Fashuai Wu
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 N. University Avenue, Ann Arbor, MI 48109-1078, USA; Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peter Ma
- Department of Biologic and Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, 1011 N. University Avenue, Ann Arbor, MI 48109-1078, USA
| | - Renny T Franceschi
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 N. University Avenue, Ann Arbor, MI 48109-1078, USA.
| |
Collapse
|
15
|
Zha D, Rayamajhi S, Sipes J, Russo A, Pathak HB, Li K, Sardiu ME, Bantis LE, Mitra A, Puri RV, Trinidad CV, Cain BP, Isenberg BC, Coppeta J, MacLaughlan S, Godwin AK, Burdette JE. Proteomic Profiling of Fallopian Tube-Derived Extracellular Vesicles Using a Microfluidic Tissue-on-Chip System. Bioengineering (Basel) 2023; 10:423. [PMID: 37106610 PMCID: PMC10135590 DOI: 10.3390/bioengineering10040423] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
The human fallopian tube epithelium (hFTE) is the site of fertilization, early embryo development, and the origin of most high-grade serous ovarian cancers (HGSOCs). Little is known about the content and functions of hFTE-derived small extracellular vesicles (sEVs) due to the limitations of biomaterials and proper culture methods. We have established a microfluidic platform to culture hFTE for EV collection with adequate yield for mass spectrometry-based proteomic profiling, and reported 295 common hFTE sEV proteins for the first time. These proteins are associated with exocytosis, neutrophil degranulation, and wound healing, and some are crucial for fertilization processes. In addition, by correlating sEV protein profiles with hFTE tissue transcripts characterized using GeoMx® Cancer Transcriptome Atlas, spatial transcriptomics analysis revealed cell-type-specific transcripts of hFTE that encode sEVs proteins, among which, FLNA, TUBB, JUP, and FLNC were differentially expressed in secretory cells, the precursor cells for HGSOC. Our study provides insights into the establishment of the baseline proteomic profile of sEVs derived from hFTE tissue, and its correlation with hFTE lineage-specific transcripts, which can be used to evaluate whether the fallopian tube shifts its sEV cargo during ovarian cancer carcinogenesis and the role of sEV proteins in fallopian tube reproductive functions.
Collapse
Affiliation(s)
- Didi Zha
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Sagar Rayamajhi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jared Sipes
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Angela Russo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Harsh B. Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Kailiang Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Mihaela E. Sardiu
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Leonidas E. Bantis
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Amrita Mitra
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Rajni V. Puri
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Camille V. Trinidad
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Brian P. Cain
- Charles Stark Draper Laboratory, Cambridge, MA 02139, USA
| | | | | | - Shannon MacLaughlan
- Department of Obstetrics and Gynecology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Andrew K. Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Joanna E. Burdette
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
16
|
Ren L, Ren Q, Wang J, He Y, Deng H, Wang X, Liu C. miR-199a-3p promotes gastric cancer progression by promoting its stemness potential via DDR2 mediation. Cell Signal 2023; 106:110636. [PMID: 36813149 DOI: 10.1016/j.cellsig.2023.110636] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 02/04/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023]
Abstract
BACKGROUND Peritoneal metastasis (PM) is an independent prognostic factor in gastric cancer (GC), however, the underlying mechanisms of PM occurrence remain unclear. METHOD The roles of DDR2 were investigated in GC and its potential relationship to PM, and orthotopic implants into nude mice were performed to assess the biological effects of DDR2 on PM. RESULTS Herein, DDR2 level is more significantly observed to elevate in PM lesion than the primary lesion. GC with DDR2-high expression evokes a worse overall survival (OS) in TCGA, similar results of the gloomy OS with high DDR2 levels are clarified via the stratifying stage of TNM. The conspicuously increased expression of DDR2 was found in GC cell lines, luciferase reporter assays verified that miR-199a-3p directly targeted DDR2 gene, which was correlated to tumor progression. We ulteriorly observed DDR2 participated in GC stemness maintenance via mediating pluripotency factor SOX2 expression and implicated in autophagy and DNA damage of cancer stem cells (CSCs). In particular, DDR2 dominated EMT programming through recruiting NFATc1-SOX2 complex to Snai1 in governing cell progression, controlling by DDR2-mTOR-SOX2 axis in SGC-7901 CSCs. Furthermore, DDR2 promoted the tumor peritoneal dissemination in gastric xenograft mouse model. CONCLUSION Phenotype screens and disseminated verifications incriminating in GC exposit the miR-199a-3p-DDR2-mTOR-SOX2 axis as a clinically actionable target for tumor PM progression. The herein-reported DDR2-based underlying axis in GC represents novel and potent tools for studying the mechanisms of PM.
Collapse
Affiliation(s)
- Lei Ren
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Qiang Ren
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jianmei Wang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yonghong He
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Hong Deng
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xing Wang
- Inflammation and Allergic Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chunfeng Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Inflammation and Allergic Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-University, Thalkirchner Str. 36, Munich 80336, Germany.
| |
Collapse
|