1
|
Zhang F, Ma Y, Li D, Wei J, Chen K, Zhang E, Liu G, Chu X, Liu X, Liu W, Tian X, Yang Y. Cancer associated fibroblasts and metabolic reprogramming: unraveling the intricate crosstalk in tumor evolution. J Hematol Oncol 2024; 17:80. [PMID: 39223656 PMCID: PMC11367794 DOI: 10.1186/s13045-024-01600-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Metabolic reprogramming provides tumors with an energy source and biofuel to support their survival in the malignant microenvironment. Extensive research into the intrinsic oncogenic mechanisms of the tumor microenvironment (TME) has established that cancer-associated fibroblast (CAFs) and metabolic reprogramming regulates tumor progression through numerous biological activities, including tumor immunosuppression, chronic inflammation, and ecological niche remodeling. Specifically, immunosuppressive TME formation is promoted and mediators released via CAFs and multiple immune cells that collectively support chronic inflammation, thereby inducing pre-metastatic ecological niche formation, and ultimately driving a vicious cycle of tumor proliferation and metastasis. This review comprehensively explores the process of CAFs and metabolic regulation of the dynamic evolution of tumor-adapted TME, with particular focus on the mechanisms by which CAFs promote the formation of an immunosuppressive microenvironment and support metastasis. Existing findings confirm that multiple components of the TME act cooperatively to accelerate the progression of tumor events. The potential applications and challenges of targeted therapies based on CAFs in the clinical setting are further discussed in the context of advancing research related to CAFs.
Collapse
Affiliation(s)
- Fusheng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Yongsu Ma
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Dongqi Li
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Jianlei Wei
- Key laboratory of Microecology-immune Regulatory Network and Related Diseases School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, 154007, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research, Peking University Health Science Center, Beijing, 100191, China
| | - Kai Chen
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Enkui Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Guangnian Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Xiangyu Chu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Xinxin Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Weikang Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Xiaodong Tian
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China.
| | - Yinmo Yang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
2
|
Dai S, Liu Y, Liu Z, Li R, Luo F, Li Y, Dai L, Peng X. Cancer-associated fibroblasts mediate resistance to anti-EGFR therapies in cancer. Pharmacol Res 2024; 206:107304. [PMID: 39002870 DOI: 10.1016/j.phrs.2024.107304] [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: 05/16/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Over the last decade, epidermal growth factor receptor (EGFR)-targeted therapies have transformed the treatment landscape for patients with advanced solid tumors. Despite these advances, resistance to anti-EGFR therapies is still a significant clinical challenge. While cell-autonomous mechanisms of resistance are well-documented, they do not fully elucidate the complexity of drug resistance. Cancer-associated fibroblasts (CAFs), key mediators within the tumor microenvironment (TME), have emerged as pivotal players in cancer progression and chemoresistance. Recent evidence implicates CAFs in resistance to anti-EGFR therapies, suggesting they may undermine treatment efficacy. This review synthesizes current data, highlighting the critical role of CAFs in resistance pathogenesis and summarizing recent therapeutic strategies targeting CAFs. We underscore the challenges and advocate for the exploration of CAFs as a potential dual-targeted approach.
Collapse
Affiliation(s)
- Shuang Dai
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yingtong Liu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610041, China
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu Sichuan, China
| | - Ruidan Li
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu Sichuan, China
| | - Feng Luo
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Lei Dai
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu Sichuan, China.
| |
Collapse
|
3
|
Wang X, Pan C, Zheng L, Wang J, Zou Q, Sun P, Zhou K, Zhao A, Cao Q, He W, Wang Y, Cheng R, Yao Z, Zhang S, Zhang H, Li M. ADAM17 variant causes hair loss via ubiquitin ligase TRIM47-mediated degradation. JCI Insight 2024; 9:e177588. [PMID: 38771644 PMCID: PMC11383180 DOI: 10.1172/jci.insight.177588] [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: 11/14/2023] [Accepted: 05/15/2024] [Indexed: 05/23/2024] Open
Abstract
Hypotrichosis is a genetic disorder characterized by a diffuse and progressive loss of scalp and/or body hair. Nonetheless, the causative genes for several affected individuals remain elusive, and the underlying mechanisms have yet to be fully elucidated. Here, we discovered a dominant variant in a disintegrin and a metalloproteinase domain 17 (ADAM17) gene caused hypotrichosis with woolly hair. Adam17 (p.D647N) knockin mice mimicked the hair abnormality in patients. ADAM17 (p.D647N) mutation led to hair follicle stem cell (HFSC) exhaustion and caused abnormal hair follicles, ultimately resulting in alopecia. Mechanistic studies revealed that ADAM17 binds directly to E3 ubiquitin ligase tripartite motif-containing protein 47 (TRIM47). ADAM17 variant enhanced the association between ADAM17 and TRIM47, leading to an increase in ubiquitination and subsequent degradation of ADAM17 protein. Furthermore, reduced ADAM17 protein expression affected the Notch signaling pathway, impairing the activation, proliferation, and differentiation of HFSCs during hair follicle regeneration. Overexpression of Notch intracellular domain rescued the reduced proliferation ability caused by Adam17 variant in primary fibroblast cells.
Collapse
Affiliation(s)
- Xiaoxiao Wang
- Department of Dermatology, Xinhua Hospital, and
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chaolan Pan
- Department of Dermatology, Xinhua Hospital, and
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Luyao Zheng
- Department of Dermatology, Xinhua Hospital, and
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Anhui Provincial Children's Hospital, Hefei, China
| | - Jianbo Wang
- Department of Dermatology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, and Henan University People's Hospital, Zhengzhou, China
| | - Quan Zou
- Department of Dermatology, Xinhua Hospital, and
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Peiyi Sun
- Department of Dermatology, Xinhua Hospital, and
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kaili Zhou
- Department of Dermatology, Xinhua Hospital, and
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Anqi Zhao
- Department of Dermatology, Xinhua Hospital, and
- Department of Dermatology, The Children's Hospital of Fudan University, Shanghai, China
| | - Qiaoyu Cao
- Department of Dermatology, Xinhua Hospital, and
- Department of Dermatology, The Children's Hospital of Fudan University, Shanghai, China
| | - Wei He
- Department of Dermatology, The Children's Hospital of Fudan University, Shanghai, China
| | - Yumeng Wang
- Department of Dermatology, Xinhua Hospital, and
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ruhong Cheng
- Department of Dermatology, Xinhua Hospital, and
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhirong Yao
- Department of Dermatology, Xinhua Hospital, and
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hui Zhang
- Department of Dermatology, Xinhua Hospital, and
| | - Ming Li
- Department of Dermatology, Xinhua Hospital, and
- Department of Dermatology, The Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
4
|
Le X, Fan YF. ADAM17 regulates the proliferation and extracellular matrix of keloid fibroblasts by mediating the EGFR/ERK signaling pathway. J Plast Surg Hand Surg 2023; 57:129-136. [PMID: 34978504 DOI: 10.1080/2000656x.2021.2017944] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
To investigate the role of a disintegrin and metalloprotease protein 17 (ADAM17) in regulating the proliferation and extracellular matrix (ECM) expression of keloid fibroblasts (KFs) via the epidermal growth factor receptor (EGFR)/extracellular signal-regulated kinase (ERK) pathway. ADAM17 expression in keloid tissues was detected by western blotting. KFs were isolated, cultured and divided into the control, shNC (negative control), shADAM17, transforming growth factor-β1 (TGF-β1), TGF-β1 + shNC and TGF-β1 + shADAM17 groups. The expression of ECM was detected by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). Western blotting was performed to detect the expression of proteins. Cell proliferation was detected by a 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay, while cell invasion and migration were examined by Transwell and wound healing assays. The expression of ADAM17 was increased in keloid tissues and KFs. Compared with the control group, the expression of p-EGFR and p-ERK/1/2/ERK1/2, as well as the expression of collagen I, collagen III, connective tissue growth factor (CTGF) and α-smooth muscle actin (α-SMA), were decreased in KFs from the shADAM17 group, with decreased cell proliferation, invasion and migration. In contrast, the TGF-β1 group presented the opposite trend in these aspects. In addition, compared with the TGF-β1 group, KFs from the TGF-β1 + shADAM17 group had decreased ECM expression, proliferation, invasion and migration. ADAM17 expression was upregulated in keloid tissues. Silencing ADAM17 may inhibit the activity of the EGFR/ERK pathway to limit the deposition of ECM in KFs with reduced proliferation, invasion and migration.
Collapse
Affiliation(s)
- Xin Le
- Department of Burn, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - You-Fen Fan
- Department of Burn, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|
5
|
Wang K, Xuan Z, Liu X, Zheng M, Yang C, Wang H. Immunomodulatory role of metalloproteinase ADAM17 in tumor development. Front Immunol 2022; 13:1059376. [PMID: 36466812 PMCID: PMC9715963 DOI: 10.3389/fimmu.2022.1059376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/03/2022] [Indexed: 12/25/2023] Open
Abstract
ADAM17 is a member of the a disintegrin and metalloproteinase (ADAM) family of transmembrane proteases involved in the shedding of some cell membrane proteins and regulating various signaling pathways. More than 90 substrates are regulated by ADAM17, some of which are closely relevant to tumor formation and development. Besides, ADAM17 is also responsible for immune regulation and its substrate-mediated signal transduction. Recently, ADAM17 has been considered as a major target for the treatment of tumors and yet its immunomodulatory roles and mechanisms remain unclear. In this paper, we summarized the recent understanding of structure and several regulatory roles of ADAM17. Importantly, we highlighted the immunomodulatory roles of ADAM17 in tumor development, as well as small molecule inhibitors and monoclonal antibodies targeting ADAM17.
Collapse
Affiliation(s)
- Kai Wang
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Zixue Xuan
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xiaoyan Liu
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Meiling Zheng
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Chao Yang
- National Engineering Research Center for Marine Aquaculture, Institute of Innovation & Application, Zhejiang Ocean University, Zhoushan, China
| | - Haiyong Wang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
6
|
ADAM10 and ADAM17 as Biomarkers Linked to Inflammation, Metabolic Disorders and Colorectal Cancer. Curr Issues Mol Biol 2022; 44:4517-4527. [PMID: 36286024 PMCID: PMC9600049 DOI: 10.3390/cimb44100309] [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: 08/29/2022] [Revised: 09/15/2022] [Accepted: 09/22/2022] [Indexed: 11/18/2022] Open
Abstract
ADAM10 and ADAM17 have a role in inflammation and diseases associated with inflammation, such as diabetes, cardiovascular diseases (CVD) or cancer, e.g., colorectal cancer (CRC). The aim of this study was to evaluate whether ADAM10 and ADAM17 could be biomarkers of CRC. To achieve this goal, CRC tumors and a surgical margin from 72 patients with CRC were collected. The concentration of ADAM proteins was measured by the ELISA method. Results were analyzed statistically and compared with selected clinical parameters. We found that ADAM17 protein concentration in the tumor samples was higher in patients with diabetes mellitus type 2 (DMT2) (0.28 vs. 0.2 ng/µg protein; p = 0.01) and in the surgical margin was higher both in patients with coexisting DMT2 (0.22 vs. 0.16 ng/µg protein; p < 0.05) and CVD (0.21 vs. 0.13 ng/µg protein; p < 0.01). The concentration of ADAM10 was higher in the surgical margin than in the tumor (249.34 vs. 228.82 pg/µg protein), and the concentration of ADAM17 was higher in the tumor than in the margin (0.23 vs. 0.18 ng/µg protein), but results were not statistically significant. In conclusion, the results of our study indicate that ADAM10 and ADAM17 may be potential biomarkers in cancer linked with DMT2 and CVD as diseases associated with inflammation.
Collapse
|
7
|
Łukaszewicz-Zając M, Pączek S, Mroczko B. A Disintegrin and Metalloproteinase (ADAM) Family-Novel Biomarkers of Selected Gastrointestinal (GI) Malignancies? Cancers (Basel) 2022; 14:cancers14092307. [PMID: 35565436 PMCID: PMC9101749 DOI: 10.3390/cancers14092307] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 02/06/2023] Open
Abstract
The global burden of gastrointestinal (GI) cancers is expected to increase. Therefore, it is vital that novel biomarkers useful for the early diagnosis of these malignancies are established. A growing body of data has linked secretion of proteolytic enzymes, such as metalloproteinases (MMPs), which destroy the extracellular matrix, to pathogenesis of GI tumours. A disintegrin and metalloproteinase (ADAM) proteins belong to the MMP family but have been proven to be unique due to both proteolytic and adhesive properties. Recent investigations have demonstrated that the expression of several ADAMs is upregulated in GI cancer cells. Thus, the objective of this review is to present current findings concerning the role of ADAMs in the pathogenesis of GI cancers, particularly their involvement in the development and progression of colorectal, pancreatic and gastric cancer. Furthermore, the prognostic significance of selected ADAMs in patients with GI tumours is also presented. It has been proven that ADAM8, 9, 10, 12, 15, 17 and 28 might stimulate the proliferation and invasion of GI malignancies and may be associated with unfavourable survival. In conclusion, this review confirms the role of selected ADAMs in the pathogenesis of the most common GI cancers and indicates their promising significance as potential prognostic biomarkers as well as therapeutic targets for GI malignancies. However, due to their non-specific nature, future research on ADAM biology should be performed to elucidate new strategies for the diagnosis of these common and deadly malignancies and treatment of patients with these diseases.
Collapse
Affiliation(s)
- Marta Łukaszewicz-Zając
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland;
- Correspondence:
| | - Sara Pączek
- Department of Biochemical Diagnostics, University Hospital of Bialystok, 15-269 Bialystok, Poland;
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland;
- Department of Neurodegeneration Diagnostics, Medical University, 15-269 Bialystok, Poland
| |
Collapse
|
8
|
Li Q, Zhu B, Chen J, Wang H, Wu Y, Chen H, He X. Effects of Oncogene Neuregulin 1 on Breast Cancer Cells. Pak J Biol Sci 2022; 25:345-352. [PMID: 35638529 DOI: 10.3923/pjbs.2022.345.352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
<b>Background and Objectives:</b> The NRG1 fusion protein is a driving factor for the occurrence and development of many tumours. We aimed to evaluate the effects of oncogene Neuregulin 1 (NRG1) on the proliferation and migration of breast cancer cells. <b>Materials and Methods:</b> Target gene NRG1 was transfected into breast cancer cells using the gene transfection technique and the migration ability of cells was observed by wound healing assay. The migration and invasion abilities of cells were further observed by Transwell assay and cell apoptosis was observed by TUNEL staining. The cell cycle distribution of breast cancer cells was detected by flow cytometry. <b>Results:</b> The wound healing assay exhibited that breast cancer cells overexpressing NRG1 exhibited stronger migration (p = 0.0047). More breast cancer cells of up-regulating NRG1 penetrated the transwell chamber, showing enhanced invasion ability (p = 0.0029). The TUNEL assay and flow cytometry demonstrated that NRG1 inhibited cell apoptosis and made them enter the active division stage. <b>Conclusion:</b> The NRG1 can promote the malignant function of breast cancer cells by augmenting migration and invasion abilities. High expression of NRG1 remarkably suppressed the apoptosis of breast cancer cells.
Collapse
|
9
|
ADAM17-A Potential Blood-Based Biomarker for Detection of Early-Stage Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13215563. [PMID: 34771725 PMCID: PMC8583642 DOI: 10.3390/cancers13215563] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 01/11/2023] Open
Abstract
Simple Summary Ovarian cancer has the highest lethality among gynecological tumors. Therefore, it is essential to find reliable biomarkers to improve early detection. This is the first report describing ADAM17 detection in serum and ascites fluid of ovarian cancer patients. A high ADAM17 concentration in serum at primary diagnosis is associated with early FIGO stages and predicts complete resection of the tumor mass. In addition, ADAM17 and CA-125 complement each other, especially in the diagnosis of early stages. In summary, ADAM17 appears to be a promising screening marker for detecting early-stage ovarian cancer. Abstract Ovarian cancer has the highest mortality rate among gynecological tumors. This is based on late diagnosis and the lack of early symptoms. To improve early detection, it is essential to find reliable biomarkers. The metalloprotease ADAM17 could be a potential marker, as it is highly expressed in many solid tumors, including ovarian and breast cancer. The aim of this work is to evaluate the relevance of ADAM17 as a potential diagnostic blood-based biomarker in ovarian cancer. Ovarian cancer cell lines IGROV-1 and A2780, as well as primary patient-derived tumor cells obtained from tumor tissue and ascitic fluid, were cultured to analyze ADAM17 abundance in the culture supernatant. In a translational approach, a cohort of 117 well-characterized ovarian cancer patients was assembled and ADAM17 levels in serum and corresponding ascitic fluid were determined at primary diagnosis. ADAM17 was quantified by enzyme-linked immunosorbent assay (ELISA). In the present study, ADAM17 was detected in the culture supernatant of ovarian cancer cell lines and primary cells. In addition, ADAM17 was found in serum and ascites of ovarian cancer patients. ADAM17 level was significantly increased in ovarian cancer patients compared to an age-matched control group (p < 0.0001). Importantly early FIGO I/II stages, which would not have been detected by CA-125, were associated with higher ADAM17 concentrations (p = 0.007). This is the first study proposing ADAM17 as a serum tumor marker in the setting of a gynecological tumor disease. Usage of ADAM17 in combination with CA-125 and other markers could help detect early stages of ovarian cancer.
Collapse
|
10
|
Adamalizyny jako potencjalne biomarkery w wybranych nowotworach złośliwych przewodu pokarmowego. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstrakt
Nowotwory złośliwe przewodu pokarmowego zajmują czołowe miejsce zarówno wśród przyczyn zachorowań jak i zgonów z powodu chorób nowotworowych na świecie. Wciąż poszukuje się potencjalnych biomarkerów, które mogłyby posłużyć jako czynniki predykcyjne i prognostyczne w tych nowotworach. Wśród białek, które mogłyby pełnić taką rolę, wymienia się adamalizyny. Liczne białka z tej rodziny są zaangażowane w wielu etapach nowotworzenia, od procesu różnicowania się pojedynczych komórek, wzrost i progresję guza do tworzenia przerzutów odległych. Dzieje się to m.in. poprzez ścieżki sygnałowe związane z aktywacją insulinopodobnych czynników wzrostu, naskórkowych czynników wzrostu czy oddziaływanie na czynnik martwicy nowotworu TNF-α. Szczególnie istotna w wyjaśnieniu patomechanizmu rozwoju raków gruczołowych przewodu pokarmowego wydaje się ścieżka sygnałowa związana z aktywacją cytokin prozapalnych. Przewlekły stan zapalny jest bowiem dobrze udokumentowanym czynnikiem ryzyka rozwoju tej grupy chorób nowotworowych.
Poznanie roli białek z rodziny adamalizyn w rozwoju i patogenezie nowotworów złośliwych przewodu pokarmowego wymaga wciąż dalszych badań. W artykule podjęto próbę syntezy aktualnej wiedzy na temat wykorzystania wybranych białek z rodziny adamalizyn jako biomarkerów nowotworów złośliwych przewodu pokarmowego.
Collapse
|
11
|
Lim H, Koh M, Jin H, Bae M, Lee SY, Kim KM, Jung J, Kim HJ, Park SY, Kim HS, Moon WK, Hwang S, Cho NH, Moon A. Cancer-associated fibroblasts induce an aggressive phenotypic shift in non-malignant breast epithelial cells via interleukin-8 and S100A8. J Cell Physiol 2021; 236:7014-7032. [PMID: 33748944 DOI: 10.1002/jcp.30364] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/26/2021] [Accepted: 03/08/2021] [Indexed: 12/15/2022]
Abstract
Cancer-associated fibroblasts (CAFs) in the tumor microenvironment have been associated with tumor progression in breast cancer. Although crosstalk between breast cancer cells and CAFs has been studied, the effect of CAFs on non-neoplastic breast epithelial cells is not fully understood to date. Here, we investigated the effect of CAFs on aggressive phenotypes in non-neoplastic MCF10A breast epithelial cells. CAFs induced epithelial-to-mesenchymal transition (EMT) and invasive phenotype in MCF10A cells. S100A8, a potential prognostic marker in several cancers, was markedly increased in MCF10A cells by CAFs. S100A8 was crucial for CAFs-induced invasive phenotype of MCF10A cells. Among cytokines increased by CAFs, interleukin (IL)-8 induced S100A8 through transcription factors p65 NF-κB and C/EBPβ. In a xenograft mouse model with MCF10A cells and CAFs, tumor was not developed, suggesting that coinjection with CAFs may not be sufficient for in vivo tumorigenicity of MCF10A cells. Xenograft mouse tumor models with MDA-MB-231 breast carcinoma cells provided an in vivo evidence for the effect of CAFs on breast cancer progression as well as a crucial role of IL-8 in tumor growth and S100A8 expression in vivo. Using a tissue microarray of human breast cancer, we showed that S100A8 expression was correlated with poor outcomes. S100A8 expression was more frequently detected in cancer-adjacent normal human breast tissues than in normal breast tissues. Together, this study elucidated a novel mechanism for the acquisition of invasive phenotype of non-neoplastic breast cells induced by CAFs, suggesting that targeting IL-8 and S100A8 may be an effective strategy against breast cancer.
Collapse
Affiliation(s)
- Hyesol Lim
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Minsoo Koh
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Hao Jin
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Mijeong Bae
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Seung-Yeon Lee
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Kyoung Mee Kim
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Joohee Jung
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Hyun Jeong Kim
- Department of Pathology, Seoul National University Bundang Hospital, Gyeonggi, Korea
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Gyeonggi, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Hoe Suk Kim
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Woo Kyung Moon
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Sejin Hwang
- Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, Korea
| | - Nam Hoon Cho
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Aree Moon
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| |
Collapse
|
12
|
ADAM 17 and Epithelial-to-Mesenchymal Transition: The Evolving Story and Its Link to Fibrosis and Cancer. J Clin Med 2021; 10:jcm10153373. [PMID: 34362154 PMCID: PMC8347979 DOI: 10.3390/jcm10153373] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/19/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
For decades, metalloproteinase 17 (ADAM17) has been the goal of wide investigation. Since its discovery as the tumour necrosis factor-α convertase, it has been studied as the main drug target, especially in the context of inflammatory conditions and tumour. In fact, evidence is mounting to support a key role of ADAM17 in the induction of the proliferation, migration and progression of tumour cells and the trigger of the pro-fibrotic process during chronic inflammatory conditions; this occurs, probably, through the activation of epithelial-to-mesenchymal transition (EMT). EMT is a central morphologic conversion that occurs in adults during wound healing, tumour progression and organ fibrosis. EMT is characterised by the disassembly of cell–cell contacts, remodelling of the actin cytoskeleton and separation of cells, and generates fibroblast-like cells that express mesenchymal markers and have migratory properties. This transition is characterised by loss of epithelial proteins such as E-cadherin and the acquisition of new mesenchymal markers, including vimentin and a-smooth muscle actin. The present review discusses the current understanding of molecular mechanisms involved in ADAM17-dependent EMT in order to individuate innovative therapeutic strategies using ADAM17-related pathways.
Collapse
|
13
|
Wu F, Yang J, Liu J, Wang Y, Mu J, Zeng Q, Deng S, Zhou H. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct Target Ther 2021; 6:218. [PMID: 34108441 PMCID: PMC8190181 DOI: 10.1038/s41392-021-00641-0] [Citation(s) in RCA: 326] [Impact Index Per Article: 81.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/20/2021] [Accepted: 05/06/2021] [Indexed: 02/05/2023] Open
Abstract
To flourish, cancers greatly depend on their surrounding tumor microenvironment (TME), and cancer-associated fibroblasts (CAFs) in TME are critical for cancer occurrence and progression because of their versatile roles in extracellular matrix remodeling, maintenance of stemness, blood vessel formation, modulation of tumor metabolism, immune response, and promotion of cancer cell proliferation, migration, invasion, and therapeutic resistance. CAFs are highly heterogeneous stromal cells and their crosstalk with cancer cells is mediated by a complex and intricate signaling network consisting of transforming growth factor-beta, phosphoinositide 3-kinase/AKT/mammalian target of rapamycin, mitogen-activated protein kinase, Wnt, Janus kinase/signal transducers and activators of transcription, epidermal growth factor receptor, Hippo, and nuclear factor kappa-light-chain-enhancer of activated B cells, etc., signaling pathways. These signals in CAFs exhibit their own special characteristics during the cancer progression and have the potential to be targeted for anticancer therapy. Therefore, a comprehensive understanding of these signaling cascades in interactions between cancer cells and CAFs is necessary to fully realize the pivotal roles of CAFs in cancers. Herein, in this review, we will summarize the enormous amounts of findings on the signals mediating crosstalk of CAFs with cancer cells and its related targets or trials. Further, we hypothesize three potential targeting strategies, including, namely, epithelial-mesenchymal common targets, sequential target perturbation, and crosstalk-directed signaling targets, paving the way for CAF-directed or host cell-directed antitumor therapy.
Collapse
Affiliation(s)
- Fanglong Wu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jin Yang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Junjiang Liu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ye Wang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jingtian Mu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Qingxiang Zeng
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shuzhi Deng
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hongmei Zhou
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
14
|
Abstract
Secretory proteins in tumor tissues are important components of the tumor microenvironment. Secretory proteins act on tumor cells or stromal cells or mediate interactions between tumor cells and stromal cells, thereby affecting tumor progression and clinical treatment efficacy. In this paper, recent research advances in secretory proteins in malignant tumors are reviewed.
Collapse
Affiliation(s)
- Na Zhang
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jiajie Hao
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yan Cai
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Mingrong Wang
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
15
|
Strategies to Target ADAM17 in Disease: From its Discovery to the iRhom Revolution. Molecules 2021; 26:molecules26040944. [PMID: 33579029 PMCID: PMC7916773 DOI: 10.3390/molecules26040944] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 02/07/2023] Open
Abstract
For decades, disintegrin and metalloproteinase 17 (ADAM17) has been the object of deep investigation. Since its discovery as the tumor necrosis factor convertase, it has been considered a major drug target, especially in the context of inflammatory diseases and cancer. Nevertheless, the development of drugs targeting ADAM17 has been harder than expected. This has generally been due to its multifunctionality, with over 80 different transmembrane proteins other than tumor necrosis factor α (TNF) being released by ADAM17, and its structural similarity to other metalloproteinases. This review provides an overview of the different roles of ADAM17 in disease and the effects of its ablation in a number of in vivo models of pathological conditions. Furthermore, here, we comprehensively encompass the approaches that have been developed to accomplish ADAM17 selective inhibition, from the newest non-zinc-binding ADAM17 synthetic inhibitors to the exploitation of iRhom2 to specifically target ADAM17 in immune cells.
Collapse
|
16
|
Cancer-associated fibroblast-secreted exosomal miR-423-5p promotes chemotherapy resistance in prostate cancer by targeting GREM2 through the TGF-β signaling pathway. Exp Mol Med 2020; 52:1809-1822. [PMID: 33144675 PMCID: PMC8080786 DOI: 10.1038/s12276-020-0431-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 12/24/2022] Open
Abstract
Therapeutic failure in prostate cancer (PC) is believed to result from its unusually invasive and metastatic nature. Cancer-associated fibroblasts (CAFs) are essential in the tumor microenvironment. We intended to study the role of CAF-derived exosomes in the context of PC and the potential regulatory mechanism associated with miR-423-5p and GREM2. CAF-derived exosomes decreased the chemosensitivity of parental PC cells and enhanced the drug resistance of drug-resistant cells. PC-associated fibroblast-derived exosomes carrying miR-423-5p increased the resistance of PC to taxane by inhibiting GREM2 through the TGF-β pathway. Inhibition of the TGF-β pathway partially reversed the increased drug resistance in PC cells induced by CAF-derived exosomes. Inhibition of miR-423-5p enhanced the drug sensitivity of PC cells in vivo. We showed that CAF-secreted exosomal miR-423-5p promoted chemotherapy resistance in PC by targeting GREM2 through the TGF-β pathway. This study may allow the development of novel approaches for PC.
Collapse
|
17
|
Stromal CCL2 Signaling Promotes Mammary Tumor Fibrosis through Recruitment of Myeloid-Lineage Cells. Cancers (Basel) 2020; 12:cancers12082083. [PMID: 32731354 PMCID: PMC7465971 DOI: 10.3390/cancers12082083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022] Open
Abstract
Obesity is correlated with breast tumor desmoplasia, leading to diminished chemotherapy response and disease-free survival. Obesity causes chronic, macrophage-driven inflammation within breast tissue, initiated by chemokine ligand 2 (CCL2) signaling from adipose stromal cells. To understand how CCL2-induced inflammation alters breast tumor pathology, we transplanted oncogenically transformed human breast epithelial cells with breast stromal cells expressing CCL2 or empty vector into murine mammary glands and examined tumor formation and progression with time. As tumors developed, macrophages were rapidly recruited, followed by the emergence of cancer-associated fibroblasts (CAFs) and collagen deposition. Depletion of CD11b + myeloid lineage cells early in tumor formation reduced tumor growth, CAF numbers, and collagen deposition. CCL2 expression within developing tumors also enhanced recruitment of myeloid progenitor cells from the bone marrow into the tumor site. The myeloid progenitor cell population contained elevated numbers of fibrocytes, which exhibited platelet-derived growth factor receptor-alpha (PDGFRα)-dependent colony formation and growth in vitro. Together, these results suggest that chronic inflammation induced by CCL2 significantly enhances tumor growth and promotes the formation of a desmoplastic stroma through early recruitment of macrophages and fibrocytes into the tumor microenvironment. Fibrocytes may be a novel target in the tumor microenvironment to reduce tumor fibrosis and enhance treatment responses for obese breast cancer patients.
Collapse
|
18
|
Ni P, Yu M, Zhang R, He M, Wang H, Chen S, Duan G. Prognostic Significance of ADAM17 for Gastric Cancer Survival: A Meta-Analysis. ACTA ACUST UNITED AC 2020; 56:medicina56070322. [PMID: 32610677 PMCID: PMC7404708 DOI: 10.3390/medicina56070322] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/14/2020] [Accepted: 06/25/2020] [Indexed: 12/24/2022]
Abstract
Background and objectives: The prognostic role of a disintegrin and metalloproteinase (ADAM) 17 has been widely assessed in gastric cancer. However, the results are inconsistent. We performed a meta-analysis to evaluate the prognostic significance of ADAM17 and its association with clinicopathological parameters. Methods: The databases of PubMed, Web of Science, and Embase were searched for relevant articles published up to April 2020. The reported hazard ratios (HRs) and odds ratios (ORs) and their corresponding 95% confidence intervals (CIs) were pooled to evaluate the strength of the association. Stata 12.1 was used to perform statistical analyses. Results: Seven studies, including 1757 patients, were screened for the meta-analysis. Compared with the high ADAM17 expression group, the pooled HR was higher in the low ADAM17 expression group (HR = 2.04, 95% CI 1.66–2.50; I2 = 18.1%; p = 0.299). High ADAM17 expression was also related to the tumor node metastasis (TNM) stages (OR = 4.09, 95% CI 1.85–9.04; I2 = 84.1%; p = 0.000), lymph node metastasis (OR = 3.08, 95% CI 1.13–8.36; I2 = 79.7%; p = 0.007), and ages (OR = 1.65, 95% CI 1.24–2.21; I2 = 0%; p = 0.692) of the gastric patients. Conclusions: This meta-analysis revealed that ADAM17 is a significant biomarker for poor prognosis in gastric cancer.
Collapse
Affiliation(s)
- Peng Ni
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, China; (P.N.); (M.Y.); (M.H.); (H.W.); (S.C.); (G.D.)
| | - Mingyang Yu
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, China; (P.N.); (M.Y.); (M.H.); (H.W.); (S.C.); (G.D.)
| | - Rongguang Zhang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, China; (P.N.); (M.Y.); (M.H.); (H.W.); (S.C.); (G.D.)
- College of Public Health, Hainan Medical University, Haikou 571199, China
- Correspondence: ; Tel.: +86-135-2558-3039; Fax: +86-371-6699-7182
| | - Mengya He
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, China; (P.N.); (M.Y.); (M.H.); (H.W.); (S.C.); (G.D.)
| | - Haiyan Wang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, China; (P.N.); (M.Y.); (M.H.); (H.W.); (S.C.); (G.D.)
| | - Shuaiyin Chen
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, China; (P.N.); (M.Y.); (M.H.); (H.W.); (S.C.); (G.D.)
| | - Guangcai Duan
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, China; (P.N.); (M.Y.); (M.H.); (H.W.); (S.C.); (G.D.)
| |
Collapse
|
19
|
Adamalysines as Biomarkers and a Potential Target of Therapy in Colorectal Cancer Patients: Preliminary Results. DISEASE MARKERS 2019; 2019:5035234. [PMID: 31565100 PMCID: PMC6745173 DOI: 10.1155/2019/5035234] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/21/2019] [Accepted: 07/24/2019] [Indexed: 01/18/2023]
Abstract
Colorectal cancer is one of the most common cancers in the world. Due to its still undetermined pathogenesis, we are searching for signaling pathways that are important in the development of colorectal cancer. In this article, we present results of study on the role of ADAM proteins in colorectal cancer. The study included 85 adult colorectal cancer patients (48 men, 37 women) and 25 patients in the control group (after diagnostic colonoscopy-without cancer). During hospitalization, a serum sample (3 cm3) was collected from the study and control group, anthropometric measurements were conducted and others clinical data were analyzed. In the serum ADAM10, 12, 17, and 28, protein concentrations were determined and, in the next step, examined the relationship between ADAMs concentrations and selected clinical parameters in both groups. The analysis showed that serum levels of ADAM10 and ADAM28 are significantly higher in patients with colorectal cancer and correlate with histopathological grading and with presence of distant metastases. Moreover, noticed the trend to correlate concentrations of adamalysines with higher BMI score. One of the functions of adamalysines is the activation of growth factors involved in cancer, including IGF and TNFα. The increased activity of adamalysines in patients may play a role in the pathogenesis of colorectal cancer. Our study highlights the prevalence of metabolic disorders in the group of patients with diagnosed CRC, and this cancer seems to be a further complication of obesity.
Collapse
|
20
|
Houthuijzen JM, Jonkers J. Cancer-associated fibroblasts as key regulators of the breast cancer tumor microenvironment. Cancer Metastasis Rev 2019; 37:577-597. [PMID: 30465162 DOI: 10.1007/s10555-018-9768-3] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tumor cells exist in close proximity with non-malignant cells. Extensive and multilayered crosstalk between tumor cells and stromal cells tailors the tumor microenvironment (TME) to support survival, growth, and metastasis. Fibroblasts are one of the largest populations of non-malignant host cells that can be found within the TME of breast, pancreatic, and prostate tumors. Substantial scientific evidence has shown that these cancer-associated fibroblasts (CAFs) are not only associated with tumors by proximity but are also actively recruited to developing tumors where they can influence other cells of the TME as well as influencing tumor cell survival and metastasis. This review discusses the impact of CAFs on breast cancer biology and highlights their heterogeneity, origin and their role in tumor progression, ECM remodeling, therapy resistance, metastasis, and the challenges ahead of targeting CAFs to improve therapy response.
Collapse
Affiliation(s)
- J M Houthuijzen
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - J Jonkers
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Düsterhöft S, Lokau J, Garbers C. The metalloprotease ADAM17 in inflammation and cancer. Pathol Res Pract 2019; 215:152410. [PMID: 30992230 DOI: 10.1016/j.prp.2019.04.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/05/2019] [Accepted: 04/05/2019] [Indexed: 12/23/2022]
Abstract
Proteolytic cleavage of transmembrane proteins is an important post-translational modification that regulates the biological function of numerous transmembrane proteins. Among the 560 proteases encoded in the human genome, the metalloprotease A Disintegrin and Metalloprotease 17 (ADAM17) has gained much attention in recent years and has emerged as a central regulatory hub in inflammation, immunity and cancer development. In order to do so, ADAM17 cleaves a variety of substrates, among them the interleukin-6 receptor (IL-6R), the pro-inflammatory cytokine tumor necrosis factor α (TNFα) and most ligands of the epidermal growth factor receptor (EGFR). This review article provides an overview of the functions of ADAM17 with a special focus on its cellular regulation. It highlights the importance of ADAM17 to understand the biology of IL-6 and TNFα and their role in inflammatory diseases. Finally, the role of ADAM17 in the formation and progression of different tumor entities is discussed.
Collapse
Affiliation(s)
- Stefan Düsterhöft
- Institute for Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Juliane Lokau
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany.
| |
Collapse
|
22
|
Kong J, Zhao H, Shang Q, Ma Z, Kang N, Tan J, Ahmed Ibrahim Alraimi H, Liu T. Establishment and characterization of a carcinoma-associated fibroblast cell line derived from a human salivary gland adenoid cystic carcinoma. ACTA ACUST UNITED AC 2018; 24:11-18. [PMID: 29734861 DOI: 10.1080/15419061.2018.1464000] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Salivary gland adenoid cystic carcinoma (SACC) is one of the most common malignancies in the oral and maxillofacial region. Carcinoma-associated fibroblast (CAF) is an important component in the tumor microenvironment and participates in SACC progression. In this study, we established a CAF cell line derived from a human SACC and named it CAF-SA. It was identified that CAF-SA expressed typical CAF biomarkers. Then, we studied the cellular communications between CAF-SA, tumor cells and endothelial cells. It was found that CAF-SA promoted the migration, invasion, and proliferation of SACC tumor cells in vitro. In addition, tube formation by endothelial cells was enhanced by CAF-SA. In vivo experiment showed that SACC cells formed larger xenografts in nude mice when they were transplanted with CAF-SA. Overall, we demonstrated that CAF-SA exhibited the most important defining feature of CAF by promoting cancer progression.
Collapse
Affiliation(s)
- Jing Kong
- a College of Stomatology , Dalian Medical University , Dalian , China
| | - Han Zhao
- a College of Stomatology , Dalian Medical University , Dalian , China
| | - Qianhui Shang
- a College of Stomatology , Dalian Medical University , Dalian , China
| | - Zhifei Ma
- a College of Stomatology , Dalian Medical University , Dalian , China
| | - Ni Kang
- a College of Stomatology , Dalian Medical University , Dalian , China
| | - Junling Tan
- a College of Stomatology , Dalian Medical University , Dalian , China
| | | | - Tingjiao Liu
- a College of Stomatology , Dalian Medical University , Dalian , China
| |
Collapse
|
23
|
Li YY, Tao YW, Gao S, Li P, Zheng JM, Zhang SE, Liang J, Zhang Y. Cancer-associated fibroblasts contribute to oral cancer cells proliferation and metastasis via exosome-mediated paracrine miR-34a-5p. EBioMedicine 2018; 36:209-220. [PMID: 30243489 PMCID: PMC6197737 DOI: 10.1016/j.ebiom.2018.09.006] [Citation(s) in RCA: 240] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/20/2018] [Accepted: 09/05/2018] [Indexed: 11/18/2022] Open
Abstract
Background Cancer-associated fibroblasts (CAFs) play an important role in regulating tumor progression by transferring exosomes to neighboring cells. Our aim was to clarify the role of microRNA encapsulated in the exosomes derived from CAFs in oral squamous cell carcinoma (OSCC). Methods We examined the microRNA expression profiles of exosomes derived from CAFs and donor-matched normal fibroblasts (NFs) from patients with OSCC. We used confocal microscopy to examine the transportation of exosomal miR-34a-5p between CAFs and OSCC cells. Next, luciferase reporter and its mutant plasmids were used to confirm direct target gene of miR-34a-5p. Phenotypic assays and in vivo tumor growth experiments were used to investigate the functional significance of exosomal miR-34a-5p. Findings We found that the expression of miR-34a-5p in CAF-derived exosomes was significantly reduced, and fibroblasts could transfer exosomal miR-34a-5p to OSCC cells. In xenograft experiments, miR-34a-5p overexpression in CAFs could inhibit the tumorigenesis of OSCC cells. We further revealed that miR-34a-5p binds to its direct downstream target AXL to suppress OSCC cell proliferation and metastasis. Stable ectopic expression of AXL in OSCC cells overexpressing miR-34a-5p restored proliferation and motility abolished by the miRNA. The miR-34a-5p/AXL axis promoted OSCC progression via the AKT/GSK-3β/β-catenin signaling pathway, which could induce the epithelial-mesenchymal transition (EMT) to promote cancer cells metastasis. The miR-34a-5p/AXL axis enhanced nuclear translocation of β-catenin and then induced transcriptional upregulation of SNAIL, which in turn activated both MMP-2 and MMP-9. Interpretation The miR-34a-5p/AXL axis confers aggressiveness in oral cancer cells through the AKT/GSK-3β/β-catenin/Snail signaling cascade and might represent a therapeutic target for OSCC. Fund National Natural Science Foundation of China. CAFs transfer miR-34a-5p-devoid exosomes to OSCC cells. Exosomal miR-34a-5p mediates the proliferation and motility of OSCC cells through regulating AXL. MiR-34a-5p/AXL axis confers aggressiveness in OSCC through AKT/GSK-3β/β-catenin/Snail signaling cascade.
Collapse
Affiliation(s)
- Yao-Yin Li
- Department of Pediatric Dentistry, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China; Guangdong Province Key Laboratory of Stomatology, Guangzhou, Guangdong 510080, China.
| | - Yi-Wei Tao
- Guangdong Province Key Laboratory of Stomatology, Guangzhou, Guangdong 510080, China; Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Shuo Gao
- Department of Pediatric Dentistry, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Pei Li
- Department of Pediatric Dentistry, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Jian-Mao Zheng
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Si-En Zhang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Jianfeng Liang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| | - Yuejiao Zhang
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, China
| |
Collapse
|
24
|
Hu B, Meng X, Zhang Y, Hossain MM, Wu L, Zhang Y, Peng X, Zhang X. Short hairpin RNA-mediated gene silencing of ADAM17 inhibits the growth of breast cancer MCF‑7 cells in vitro and in vivo and its mechanism of action. Oncol Rep 2018; 39:1640-1648. [PMID: 29393483 PMCID: PMC5868399 DOI: 10.3892/or.2018.6237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 01/09/2018] [Indexed: 12/13/2022] Open
Abstract
A disintegrin and metalloprotease 17 (ADAM17) is highly expressed in many malignant tumors and is closely related to their development. We showed in a previous study that silencing of ADAM17 by siRNA inhibited the growth of MCF-7 breast cancer cells in vitro and in vivo. In the present study, we investigated the effects of ADAM17-short hairpin RNA (ADAM17-shRNA) on MCF-7 breast cancer cells and explored the potential action pathway. In vitro, transfection of shRNAs was performed using a lentivirus, and the effects of ADAM17-shRNA on invasion, proliferation and cell cycle distribution of MCF-7 cells were assessed by Boyden chamber method, real-time cell analysis and flow cytometry, respectively. In vivo, MCF-7 cells with different administrations were transplanted subcutaneously into nude mice, and the effect of ADAM17-shRNA on the growth of transplanted tumors was assessed. In addition, the morphological structures were observed by H&E staining, and the expression of ADAM17 and Ki-67 was assessed by immunohistochemistry; expression of ADAM17, EGFR, p-EGFR, AKT, p-AKT, ERK and p-ERK proteins was assessed by western blotting, respectively. Our data showed that ADAM17-shRNA successfully inhibited ADAM17 mRNA expression, invasion and proliferation of MCF-7 cells resulting in G0/G1 phase arrest, and significantly inhibited the growth of transplanted tumors with larger areas of necrosis, low expression of ADAM17 and Ki-67 and reduced protein expression of ADAM17, EGFR, p-EGFR, AKT, p-AKT, ERK, and p-ERK in the tumor tissues. The present research suggests that ADAM17-shRNA can inhibit MCF-7 cell invasion and proliferation in vitro and inhibit MCF-7 xenograft growth in vivo through the EGFR/PI3K/AKT and EGFR/MEK/ERK signaling pathways.
Collapse
Affiliation(s)
- Baoshan Hu
- Department of Surgical Oncology, Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Xiangchao Meng
- Department of Surgical Oncology, Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Yan Zhang
- Department of Surgical Oncology, Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Mohammad Monir Hossain
- International Education College, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Lijun Wu
- Department of Surgical Oncology, Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Yuanyuan Zhang
- Department of Surgical Oncology, Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Xiaobing Peng
- Department of Surgical Oncology, Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Xuepeng Zhang
- Department of Surgical Oncology, Affiliated Hospital, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
25
|
Stromal Fibroblasts from the Interface Zone of Triple Negative Breast Carcinomas Induced Epithelial-Mesenchymal Transition and its Inhibition by Emodin. PLoS One 2017; 12:e0164661. [PMID: 28060811 PMCID: PMC5218416 DOI: 10.1371/journal.pone.0164661] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 09/28/2016] [Indexed: 12/31/2022] Open
Abstract
“Triple negative breast cancer” (TNBC) is associated with a higher rate and earlier time of recurrence and worse prognosis after recurrence. In this study, we aimed to examine the crosstalk between fibroblasts and TNBC cells. The fibroblasts were isolated from TNBC patients’ tissue in tumor burden zones, distal normal zones and interface zones. The fibroblasts were indicated as cancer-associated fibroblasts (CAFs), normal zone fibroblasts (NFs) and interface zone fibroblasts (INFs). Our study found that INFs grew significantly faster than NFs and CAFs in vitro. The epithelial BT20 cells cultured with the conditioned medium of INFs (INFs-CM) and CAFs (CAFs-CM) showed more spindle-like shape and cell scattering than cultured with the conditioned medium of NFs (NFs-CM). These results indicated that factors secreted by INFs-CM or CAFs-CM could induce the epithelial-mesenchymal transition (EMT) phenotype in BT20 cells. Using an in vitro co-culture model, INFs or CAFs induced EMT and promoted cancer cell migration in BT20 cells. Interestingly, we found that emodin inhibited INFs-CM or CAFs-CM-induced EMT programming and phenotype in BT20 cells. Previous studies reported that CAFs and INFs-secreted TGF-β promoted human breast cancer cell proliferation, here; our results indicated that TGF-β initiated EMT in BT20 cells. Pretreatment with emodin significantly suppressed the TGF-β-induced EMT and cell migration in BT20 cells. These results suggest that emodin may be used as a novel agent for the treatment of TNBC.
Collapse
|
26
|
Carnet O, Lecomte J, Masset A, Primac I, Durré T, Maertens L, Detry B, Blacher S, Gilles C, Péqueux C, Paupert J, Foidart JM, Jerusalem G, Cataldo D, Noel A. Mesenchymal Stem Cells Shed Amphiregulin at the Surface of Lung Carcinoma Cells in a Juxtacrine Manner. Neoplasia 2016; 17:552-63. [PMID: 26297433 PMCID: PMC4547406 DOI: 10.1016/j.neo.2015.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 06/23/2015] [Accepted: 07/02/2015] [Indexed: 12/13/2022] Open
Abstract
Solid tumors comprise cancer cells and different supportive stromal cells, including mesenchymal stem cells (MSCs), which have recently been shown to enhance tumor growth and metastasis. We provide new mechanistic insights into how bone marrow (BM)–derived MSCs co-injected with Lewis lung carcinoma cells promote tumor growth and metastasis in mice. The proinvasive effect of BM-MSCs exerted on tumor cells relies on an unprecedented juxtacrine action of BM-MSC, leading to the trans-shedding of amphiregulin (AREG) from the tumor cell membrane by tumor necrosis factor-α–converting enzyme carried by the BM-MSC plasma membrane. The released soluble AREG activates cancer cells and promotes their invasiveness. This novel concept is supported by the exploitation of different 2D and 3D culture systems and by pharmacological approaches using a tumor necrosis factor-α–converting enzyme inhibitor and AREG-blocking antibodies. Altogether, we here assign a new function to BM-MSC in tumor progression and establish an uncovered link between AREG and BM-MSC.
Collapse
Affiliation(s)
- Oriane Carnet
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium
| | - Julie Lecomte
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium
| | - Anne Masset
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium
| | - Irina Primac
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium
| | - Tania Durré
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium
| | - Ludovic Maertens
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium
| | - Benoit Detry
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium
| | - Silvia Blacher
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium
| | - Christine Gilles
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium
| | - Christel Péqueux
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium
| | - Jenny Paupert
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium
| | - Jean-Michel Foidart
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium
| | - Guy Jerusalem
- Department of Medical Oncology, Centre Hospitalier Universitaire (CHU), Sart Tilman, B-4000 Liège, Belgium
| | - Didier Cataldo
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium
| | - Agnès Noel
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, B-4000 Liège, Belgium.
| |
Collapse
|
27
|
Abstract
Metastasis is the underlying cause of death for the majority of breast cancer patients. Despite significant advances in recent years in basic research and clinical development, therapies that specifically target metastatic breast cancer remain inadequate, and represents the single greatest obstacle to reducing mortality of late-stage breast cancer. Recent efforts have leveraged genomic analysis of breast cancer and molecular dissection of tumor-stromal cross-talk to uncover a number of promising candidates for targeted treatment of metastatic breast cancer. Rational combinations of therapeutic agents targeting tumor-intrinsic properties and microenvironmental components provide a promising strategy to develop precision treatments with higher specificity and less toxicity. In this review, we discuss the emerging therapeutic targets in breast cancer metastasis, from tumor-intrinsic pathways to those that involve the host tissue components, including the immune system.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, United States
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, United States.
| |
Collapse
|
28
|
Xiong Y, McDonald LT, Russell DL, Kelly RR, Wilson KR, Mehrotra M, Soloff AC, LaRue AC. Hematopoietic stem cell-derived adipocytes and fibroblasts in the tumor microenvironment. World J Stem Cells 2015; 7:253-265. [PMID: 25815113 PMCID: PMC4369485 DOI: 10.4252/wjsc.v7.i2.253] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 11/08/2014] [Accepted: 12/01/2014] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment (TME) is complex and constantly evolving. This is due, in part, to the crosstalk between tumor cells and the multiple cell types that comprise the TME, which results in a heterogeneous population of tumor cells and TME cells. This review will focus on two stromal cell types, the cancer-associated adipocyte (CAA) and the cancer-associated fibroblast (CAF). In the clinic, the presence of CAAs and CAFs in the TME translates to poor prognosis in multiple tumor types. CAAs and CAFs have an activated phenotype and produce growth factors, inflammatory factors, cytokines, chemokines, extracellular matrix components, and proteases in an accelerated and aberrant fashion. Through this activated state, CAAs and CAFs remodel the TME, thereby driving all aspects of tumor progression, including tumor growth and survival, chemoresistance, tumor vascularization, tumor invasion, and tumor cell metastasis. Similarities in the tumor-promoting functions of CAAs and CAFs suggest that a multipronged therapeutic approach may be necessary to achieve maximal impact on disease. While CAAs and CAFs are thought to arise from tissues adjacent to the tumor, multiple alternative origins for CAAs and CAFs have recently been identified. Recent studies from our lab and others suggest that the hematopoietic stem cell, through the myeloid lineage, may serve as a progenitor for CAAs and CAFs. We hypothesize that the multiple origins of CAAs and CAFs may contribute to the heterogeneity seen in the TME. Thus, a better understanding of the origin of CAAs and CAFs, how this origin impacts their functions in the TME, and the temporal participation of uniquely originating TME cells may lead to novel or improved anti-tumor therapeutics.
Collapse
|
29
|
Park SY, Kim HM, Koo JS. Differential expression of cancer-associated fibroblast-related proteins according to molecular subtype and stromal histology in breast cancer. Breast Cancer Res Treat 2015; 149:727-41. [PMID: 25667103 DOI: 10.1007/s10549-015-3291-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 01/28/2015] [Indexed: 12/11/2022]
Abstract
The purpose of this study aimed to investigate the clinicopathologic characteristics of breast cancer according to its cancer-associated fibroblast (CAF) phenotype. Immunohistochemistry staining of estrogen receptor, progesterone receptor, human epidermal growth factor receptor 2 (HER-2), Ki-67, podoplanin, prolyl 4-hydroxylase, fibroblast activation protein alpha (FAPα), S100A4, platelet-derived growth factor receptor alpha (PDGFRα), PDGFRβ, and chondroitin sulfate proteoglycan (NG2) was performed on tissue microarray consisting of 642 breast cancer cases. Samples were categorized into luminal A, luminal B, HER-2, or triple-negative breast cancer (TNBC) according to immunohistochemical results, whereas tumor stroma was classified into desmoplastic, sclerotic, normal-like, or inflammatory type based on histological findings. Expression of CAF-related proteins in the stroma differed depending on breast cancer molecular subtypes. All CAF-related protein expression was high (p < 0.05) in HER-2 type, whereas in luminal A, the expression of FAPα, PDGFα, PDGFβ, and NG2 was low, and in TNBC, the expression of podoplanin, prolyl 4-hydroxylase, and S100A4 was low. In the stromal component, CAF-related protein expression differed according to stromal phenotype (p < 0.001). The desmoplastic type showed high expression of podoplanin, prolyl 4-hydroxylase, S100A4, PDGFRα, and PDGFRβ, whereas the sclerotic type exhibited low expression of FAPα, PDGFα, PDGFβ, and NG2. The inflammatory type had high expression of FAPα and NG2 with low podoplanin, while normal-like type showed low expression of prolyl 4-hydroxylase and S100A4. Our results suggested that differential CAF-related protein expression depended on the molecular subtypes and stromal histologic features of breast cancer, indicating that in the future, this system could potentially use these markers for prognosis prediction and targeted therapy of breast cancer.
Collapse
|
30
|
Lee E, Lee SJ, Koskimaki JE, Han Z, Pandey NB, Popel AS. Inhibition of breast cancer growth and metastasis by a biomimetic peptide. Sci Rep 2014; 4:7139. [PMID: 25409905 PMCID: PMC4238022 DOI: 10.1038/srep07139] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 11/05/2014] [Indexed: 12/21/2022] Open
Abstract
Metastasis is the main cause of mortality in cancer patients. Though there are many anti-cancer drugs targeting primary tumor growth, anti-metastatic agents are rarely developed. Angiogenesis and lymphangiogenesis are crucial for cancer progression, particularly, lymphangiogenesis is pivotal for metastasis in breast cancer. Here we report that a novel collagen IV derived biomimetic peptide inhibits breast cancer growth and metastasis by blocking angiogenesis and lymphangiogenesis. The peptide inhibits blood and lymphatic endothelial cell viability, migration, adhesion, and tube formation by targeting IGF1R and Met signals. The peptide blocks MDA-MB-231 tumor growth by inhibiting tumor angiogenesis in vivo. Moreover, the peptide inhibits lymphangiogenesis in primary tumors. MDA-MB-231 tumor conditioned media (TCM) was employed to accelerate spontaneous metastasis in tumor xenografts, and the anti-metastatic activity of the peptide was tested in this model. The peptide prevents metastasis to the lungs and lymph nodes by inhibiting TCM-induced lymphangiogenesis and angiogenesis in the pre-metastatic organs. In summary, a novel biomimetic peptide inhibits breast cancer growth and metastasis by blocking angiogenesis and lymphangiogenesis in the pre-metastatic organs as well as primary tumors.
Collapse
Affiliation(s)
- Esak Lee
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Seung Jae Lee
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Jacob E Koskimaki
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Zheyi Han
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Niranjan B Pandey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Aleksander S Popel
- 1] Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States [2] Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| |
Collapse
|
31
|
Wu B, Sha L, Wang Y, Xu W, Yu Y, Feng F, Sun C, Xia L. Diagnostic and prognostic value of a disintegrin and metalloproteinase-17 in patients with gliomas. Oncol Lett 2014; 8:2616-2620. [PMID: 25364437 PMCID: PMC4214486 DOI: 10.3892/ol.2014.2582] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 06/24/2014] [Indexed: 11/08/2022] Open
Abstract
A disintegrin and metalloproteinase-17 (ADAM17) has been shown to regulate numerous proteins involved in the cell cycle, as well as tumor oncogenes. The expression pattern of ADAM17 in glioma patients, however, is unclear. In the present study, the expression pattern and prognostic significance of ADAM17 was investigated in patients with glioma. A total of 60 glioma specimens and eight normal control samples were obtained. Immunohistochemical and western blot analyses were used to examine the expression of ADAM17. In addition, the association of ADAM17 expression with the clinicopathological parameters and the survival rates of the glioma patients was analyzed. The results showed that ADAM17 was upregulated in the high-grade glioma tissues compared with that in the low-grade and normal brain tissues of the glioma patients, and that the level increased with ascending World Health Organization tumor grade (P<0.05). Furthermore, the survival rate of the patients with ADAM17-positive tumors was lower compared with the patients with ADAM17-negative tumors. These results indicated that the overexpression of ADAM17 was correlated with a high tumor grade and a poor prognosis in patients with glioma. ADAM17 may have an important oncogenic function in glioma progression, and is a potential diagnostic and therapeutic target.
Collapse
Affiliation(s)
- Bin Wu
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Longgui Sha
- Department of Neurosurgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, Shanghai 200120, P.R. China
| | - Yong Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Wei Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yang Yu
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Fang Feng
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Caixing Sun
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Liang Xia
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|