1
|
Hapeman JD, Galwa R, Carneiro CS, Nedelcu AM. In vitro evidence for the potential of EGFR inhibitors to decrease the TGF-β1-induced dispersal of circulating tumour cell clusters mediated by EGFR overexpression. Sci Rep 2024; 14:19980. [PMID: 39198539 PMCID: PMC11358385 DOI: 10.1038/s41598-024-70358-x] [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: 07/05/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
Most cancer-related deaths are due to the spread of tumour cells throughout the body-a process known as metastasis. While in the vasculature, these cells are referred to as circulating tumour cells (CTCs) and can be found as either single cells or clusters of cells (often including platelets), with the latter having the highest metastatic potential. However, the biology of CTC clusters is poorly understood, and there are no therapies that specifically target them. We previously developed an in vitro model system for CTC clusters and proposed a new extravasation model that involves cluster dissociation, adherence, and single-cell invasion in response to TGF-β1 released by platelets. Here, we investigated TGF-β1-induced gene expression changes in this model, focusing on genes for which targeted drugs are available. In addition to the upregulation of the TGF-β1 signalling pathway, we found that (i) genes in the EGF/EGFR pathway, including those coding for EGFR and several EGFR ligands, were also induced, and (ii) Erlotinib and Osimertinib, two therapeutic EGFR/tyrosine kinase inhibitors, decreased the TGF-β1-induced adherence and invasion of the CTC cluster-like line despite the line expressing wild-type EGFR. Overall, we suggest that EGFR inhibitors have the potential to decrease the dispersal of CTC clusters that respond to TGF-β1 and overexpress EGFR (irrespective of its status) and thus could improve patient survival.
Collapse
Affiliation(s)
- Jorian D Hapeman
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada
| | - Rakshit Galwa
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada
| | - Caroline S Carneiro
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada
| | - Aurora M Nedelcu
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada.
| |
Collapse
|
2
|
Radisky ES. Extracellular proteolysis in cancer: Proteases, substrates, and mechanisms in tumor progression and metastasis. J Biol Chem 2024; 300:107347. [PMID: 38718867 PMCID: PMC11170211 DOI: 10.1016/j.jbc.2024.107347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 06/02/2024] Open
Abstract
A vast ensemble of extracellular proteins influences the development and progression of cancer, shaped and reshaped by a complex network of extracellular proteases. These proteases, belonging to the distinct classes of metalloproteases, serine proteases, cysteine proteases, and aspartic proteases, play a critical role in cancer. They often become dysregulated in cancer, with increases in pathological protease activity frequently driven by the loss of normal latency controls, diminished regulation by endogenous protease inhibitors, and changes in localization. Dysregulated proteases accelerate tumor progression and metastasis by degrading protein barriers within the extracellular matrix (ECM), stimulating tumor growth, reactivating dormant tumor cells, facilitating tumor cell escape from immune surveillance, and shifting stromal cells toward cancer-promoting behaviors through the precise proteolysis of specific substrates to alter their functions. These crucial substrates include ECM proteins and proteoglycans, soluble proteins secreted by tumor and stromal cells, and extracellular domains of cell surface proteins, including membrane receptors and adhesion proteins. The complexity of the extracellular protease web presents a significant challenge to untangle. Nevertheless, technological strides in proteomics, chemical biology, and the development of new probes and reagents are enabling progress and advancing our understanding of the pivotal importance of extracellular proteolysis in cancer.
Collapse
Affiliation(s)
- Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA.
| |
Collapse
|
3
|
Kolahi Azar H, Gharibshahian M, Rostami M, Mansouri V, Sabouri L, Beheshtizadeh N, Rezaei N. The progressive trend of modeling and drug screening systems of breast cancer bone metastasis. J Biol Eng 2024; 18:14. [PMID: 38317174 PMCID: PMC10845631 DOI: 10.1186/s13036-024-00408-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
Bone metastasis is considered as a considerable challenge for breast cancer patients. Various in vitro and in vivo models have been developed to examine this occurrence. In vitro models are employed to simulate the intricate tumor microenvironment, investigate the interplay between cells and their adjacent microenvironment, and evaluate the effectiveness of therapeutic interventions for tumors. The endeavor to replicate the latency period of bone metastasis in animal models has presented a challenge, primarily due to the necessity of primary tumor removal and the presence of multiple potential metastatic sites.The utilization of novel bone metastasis models, including three-dimensional (3D) models, has been proposed as a promising approach to overcome the constraints associated with conventional 2D and animal models. However, existing 3D models are limited by various factors, such as irregular cellular proliferation, autofluorescence, and changes in genetic and epigenetic expression. The imperative for the advancement of future applications of 3D models lies in their standardization and automation. The utilization of artificial intelligence exhibits the capability to predict cellular behavior through the examination of substrate materials' chemical composition, geometry, and mechanical performance. The implementation of these algorithms possesses the capability to predict the progression and proliferation of cancer. This paper reviewed the mechanisms of bone metastasis following primary breast cancer. Current models of breast cancer bone metastasis, along with their challenges, as well as the future perspectives of using these models for translational drug development, were discussed.
Collapse
Affiliation(s)
- Hanieh Kolahi Azar
- Department of Pathology, Tabriz University of Medical Sciences, Tabriz, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maliheh Gharibshahian
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammadreza Rostami
- Division of Food Safety and Hygiene, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Food Science and Nutrition Group (FSAN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Vahid Mansouri
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Leila Sabouri
- Department of Tissue Engineering and Applied Cell Sciences, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
4
|
Chien MH, Yang YC, Ho KH, Ding YF, Chen LH, Chiu WK, Chen JQ, Tung MC, Hsiao M, Lee WJ. Cyclic increase in the ADAMTS1-L1CAM-EGFR axis promotes the EMT and cervical lymph node metastasis of oral squamous cell carcinoma. Cell Death Dis 2024; 15:82. [PMID: 38263290 PMCID: PMC10805752 DOI: 10.1038/s41419-024-06452-9] [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: 07/12/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/25/2024]
Abstract
The matrix metalloprotease A disintegrin and metalloprotease with thrombospondin motifs 1 (ADAMTS1) was reported to be involved in tumor progression in several cancer types, but its contributions appear discrepant. At present, the role of ADAMTS1 in oral squamous cell carcinoma (SCC; OSCC) remains unclear. Herein, The Cancer Genome Atlas (TCGA) database showed that ADAMTS1 transcripts were downregulated in head and neck SCC (HNSCC) tissues compared to normal tissues, but ADAMTS1 levels were correlated with poorer prognoses of HNSCC patients. In vitro, we observed that ADAMTS1 expression levels were correlated with the invasive abilities of four OSCC cell lines, HSC-3, SCC9, HSC-3M, and SAS. Knockdown of ADAMTS1 in OSCC cells led to a decrease and its overexpression led to an increase in cell-invasive abilities in vitro as well as tumor growth and lymph node (LN) metastasis in OSCC xenografts. Mechanistic investigations showed that the cyclic increase in ADAMTS1-L1 cell adhesion molecule (L1CAM) axis-mediated epidermal growth factor receptor (EGFR) activation led to exacerbation of the invasive abilities of OSCC cells via inducing epithelial-mesenchymal transition (EMT) progression. Clinical analyses revealed that ADAMTS1, L1CAM, and EGFR levels were all correlated with worse prognoses of HNSCC patients, and patients with ADAMTS1high/L1CAMhigh or EGFRhigh tumors had the shortest overall and disease-specific survival times. As to therapeutic aspects, we discovered that an edible plant-derived flavonoid, apigenin (API), drastically inhibited expression of the ADAMTS1-L1CAM-EGFR axis and reduced the ADAMTS1-triggered invasion and LN metastasis of OSCC cells in vitro and in vivo. Most importantly, API treatment significantly prolonged survival rates of xenograft mice with OSCC. In summary, ADAMTS1 may be a useful biomarker for predicting OSCC progression, and API potentially retarded OSCC progression by targeting the ADAMTS1-L1CAM-EGFR signaling pathway.
Collapse
Affiliation(s)
- Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital Taipei, Taipei, Taiwan
| | - Yi-Chieh Yang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Kuo-Hao Ho
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Fang Ding
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Otolaryngology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Li-Hsin Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wen-Kuan Chiu
- Division of Plastic Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, School of Medicine, College of Surgery, Taipei Medical University, Taipei, Taiwan
| | - Ji-Qing Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Cancer Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Min-Che Tung
- Department of Surgery, Tungs' Taichung Metro Harbor Hospital, Taichung, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Jiunn Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
5
|
Schirmer U, Schneider SA, Khromov T, Bremmer F, Schminke B, Schliephake H, Liefeith K, Brockmeyer P. Sclerostin Alters Tumor Cell Characteristics of Oral Squamous Cell Carcinoma and May Be a Key Player in Local Bone Invasion. Cells 2024; 13:137. [PMID: 38247829 PMCID: PMC10814349 DOI: 10.3390/cells13020137] [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: 12/10/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Localized jawbone invasion is a milestone in the progression of oral squamous cell carcinoma (OSCC). The factors that promote this process are not well understood. Sclerostin is known to be involved in bone metabolism and there are preliminary reports of its involvement in bone tumors and bone metastasis. To identify a possible involvement of sclerostin in the bone invasion process of OSCC, sclerostin expression was analyzed in vitro in two different human OSCC tumor cell lines by quantitative real-time polymerase chain reaction (qRT-PCR), and the effect of recombinant human (rh)-sclerostin treatment on tumor cell capabilities was evaluated using proliferation, migration, and invasion assays. Undifferentiated human mesenchymal stem cells (hMSCs) were osteogenically differentiated and co-cultured with OSCC tumor cells to demonstrate potential interactions and migration characteristics. Sclerostin expression was evaluated in clinical cases by immunohistochemistry at the OSCC-jawbone interface in a cohort of 15 patients. Sclerostin expression was detected in both OSCC tumor cell lines in vitro and was also detected at the OSCC-jawbone interface in clinical cases. Tumor cell proliferation rate, migration and invasion ability were increased by rh-sclerostin treatment. The migration rate of tumor cells co-cultured with osteogenically differentiated hMSCs was increased. The results presented are the first data suggesting a possible involvement of sclerostin in the bone invasion process of OSCC, which deserves further investigation and may be a potential approach for drug-based tumor therapy.
Collapse
Affiliation(s)
- Uwe Schirmer
- Institute for Bioprocessing and Analytical Measurement Techniques, D-37308 Heiligenstadt, Germany; (U.S.); (S.A.S.); (K.L.)
| | - Sina Allegra Schneider
- Institute for Bioprocessing and Analytical Measurement Techniques, D-37308 Heiligenstadt, Germany; (U.S.); (S.A.S.); (K.L.)
| | - Tatjana Khromov
- Department of Clinical Chemistry, University Medical Center Goettingen, D-37075 Goettingen, Germany;
| | - Felix Bremmer
- Institute of Pathology, University Medical Center Goettingen, D-37075 Goettingen, Germany;
| | - Boris Schminke
- Department of Oral and Maxillofacial Surgery, University Medical Center Goettingen, D-37075 Goettingen, Germany; (B.S.); (H.S.)
| | - Henning Schliephake
- Department of Oral and Maxillofacial Surgery, University Medical Center Goettingen, D-37075 Goettingen, Germany; (B.S.); (H.S.)
| | - Klaus Liefeith
- Institute for Bioprocessing and Analytical Measurement Techniques, D-37308 Heiligenstadt, Germany; (U.S.); (S.A.S.); (K.L.)
| | - Phillipp Brockmeyer
- Department of Oral and Maxillofacial Surgery, University Medical Center Goettingen, D-37075 Goettingen, Germany; (B.S.); (H.S.)
| |
Collapse
|
6
|
Nolan E, Kang Y, Malanchi I. Mechanisms of Organ-Specific Metastasis of Breast Cancer. Cold Spring Harb Perspect Med 2023; 13:a041326. [PMID: 36987584 PMCID: PMC10626265 DOI: 10.1101/cshperspect.a041326] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Cancer metastasis, or the development of secondary tumors in distant tissues, accounts for the vast majority of fatalities in patients with breast cancer. Breast cancer cells show a striking proclivity to metastasize to distinct organs, specifically the lung, liver, bone, and brain, where they face unique environmental pressures and a wide variety of tissue-resident cells that together create a strong barrier for tumor survival and growth. As a consequence, successful metastatic colonization is critically dependent on reciprocal cross talk between cancer cells and host cells within the target organ, a relationship that shapes the formation of a tumor-supportive microenvironment. Here, we discuss the mechanisms governing organ-specific metastasis in breast cancer, focusing on the intricate interactions between metastatic cells and specific niche cells within a secondary organ, and the remarkable adaptations of both compartments that cooperatively support cancer growth. More broadly, we aim to provide a framework for the microenvironmental prerequisites within each distinct metastatic site for successful breast cancer metastatic seeding and outgrowth.
Collapse
Affiliation(s)
- Emma Nolan
- Tumour Host Interaction laboratory, The Francis Crick Institute, NW1 1AT London, United Kingdom
- Auckland Cancer Society Research Centre, University of Auckland, Auckland 1023, New Zealand
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, New Jersey 08544, USA
| | - Ilaria Malanchi
- Tumour Host Interaction laboratory, The Francis Crick Institute, NW1 1AT London, United Kingdom
| |
Collapse
|
7
|
Simpson KE, Staikos CA, Watson KL, Moorehead RA. Loss of MXRA8 Delays Mammary Tumor Development and Impairs Metastasis. Int J Mol Sci 2023; 24:13730. [PMID: 37762032 PMCID: PMC10530983 DOI: 10.3390/ijms241813730] [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: 08/01/2023] [Revised: 08/22/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Matrix-remodeling-associated protein 8 or MXRA8 is a transmembrane protein that can bind arthritogenic alpha viruses like the Chikungunya virus and provide viral entry into cells. MXRA8 can also interact with integrin β3 and thus possibly regulate cell-cell interactions and binding to the extracellular matrix. While MXRA8 has been associated with reduced survival in patients with colorectal and renal clear cell cancers, the role of MXRA8 in breast cancer remains largely unexplored. Therefore, the aim of this research was to determine the role of MXRA8 in breast cancer by knocking out MXRA8 in the human triple-negative breast cancer cell line MDA-MB-231. The loss of MXRA8 reduced cell proliferation in vitro but had no effect on apoptosis or migration in cultured cells. However, the loss of MXRA8 significantly delayed tumor development and reduced metastatic dissemination to the lungs in a xenograft model. RNA sequencing identified three genes, ADMATS1, TIE1, and BMP2, whose expression were significantly reduced in MXRA8-knockout tumors compared to control tumors. MXRA8 staining of a human breast cancer tissue array revealed higher levels of MXRA8 in primary tumors and metastases of aggressive tumor subtypes (TNBC and HER2+) compared to less aggressive, ER+ breast cancers. Our findings demonstrate for the first time that MXRA8 regulates the progression of human TNBC possibly through influencing the interaction of tumor cells with their microenvironment.
Collapse
Affiliation(s)
| | | | | | - Roger A. Moorehead
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (K.E.S.); (C.A.S.); (K.L.W.)
| |
Collapse
|
8
|
Zhang Q, Kanyomse Q, Luo C, Mo Q, Zhao X, Wang L, Peng W, Ren G. The Prognostic Value of ADAMTS8 and Its Role as a Tumor Suppressor in Breast Cancer. Cancer Invest 2023; 41:119-132. [PMID: 36346393 DOI: 10.1080/07357907.2022.2128367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
A disintegrin-like and metalloprotease with therombospondin type1 motif 8 (ADAMTS8) plays an important role in many malignancies. However, the clinical and biological significance of ADAMTS8 in breast cancer remain unknown. In this study, the clinical data from 1066 breast cancer patients were analyzed by The Cancer Genome Atlas (TCGA) database, and were analyzed using the correlation between ADAMTS8 expression and the clinicopathological features and prognoses. The CCK-8 assay, clone formation assay, flow cytometry and Transwell assay were used to characterize the effects of ADAMTS8 on proliferation, migration and invasion of breast cancer cells. Gene set enrichment analysis (GSEA) and western blotting were used to identify the potential molecular mechanism on how ADAMTS8 exert its biological function. ADAMTS8 overexpression correlated longer overall survival (OS) and progression-free survival (PFS). ADAMTS8 was considered as an independent prognostic factor for OS. ADAMTS8 overexpression inhibited breast cancer cell proliferation, migration and invasion in vitro, and induced G2/M cell cycle arrest. ADAMTS8 was also involved in cell cycle regulation and was associated with the EGFR/Akt signaling pathway. ADAMTS8 knockdown showed the reverse effect. Together, the results showed that ADAMTS8 functioned as a tumor suppressor gene (TGS) and could be a prognostic biomarker for breast cancer.
Collapse
Affiliation(s)
- Qia Zhang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Quist Kanyomse
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chenghao Luo
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingfan Mo
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - XunPing Zhao
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Long Wang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyan Peng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Mao S, Xia A, Tao X, Ye D, Qu J, Sun M, Wei H, Li G. A pan-cancer analysis of the prognostic and immunological roles of matrix metalloprotease-1 (MMP1) in human tumors. Front Oncol 2023; 12:1089550. [PMID: 36727076 PMCID: PMC9885257 DOI: 10.3389/fonc.2022.1089550] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
Objective Cancer remains the leading killer of human health worldwide. It has been shown that matrix metalloproteinase-1(MMP1) is related to poor prognosis in cancers such as BRCA, CESC and COAD. However, systematic pan-cancer analysis about the prognostic and immunological roles of MMP1 has not been explored. Here, the purpose of this study was to investigate the prognostic and immunological roles of MMP1 in pan-cancer and confirm cancer-promoting effect in pancreatic cancer. Methods In our study, bioinformatics were first used to analyze data from multiple databases. Then, several bioinformatics tools were utilized to investigate the role of MMP1 in 33 tumor types. Finally, molecular biology experiments were carried out to prove the cancer-promoting effect of MMP1 in pancreatic cancer. Results MMP1 expression was higher in tumor tissues than in control tissues in most tumor types. High expression of MMP1 was associated with poor overall survival (OS) and disease-free survival (DFS) in some tumor types. Further analysis of MMP1 gene mutation data showed that MMP1 mutations significantly influenced the prognosis of STAD. In addition, MMP1 expression was closely related to cancer-associated fibroblast (CAFs) infiltration in a variety of cancers and played an important role on immune infiltration score, tumor mutational burden (TMB) and microsatellite instability (MSI). Gene Ontology enrichment analysis indicated that these 20 genes were mainly related to extracellular structure organization/extracellular matrix organization/extracellular matrix disassembly/collagen metabolic process in the enriched biological processes. Finally, molecular biology experiments confirmed the cancer-promoting effect of MMP1 in pancreatic cancer. Conclusions Our pan-cancer analysis comprehensively proved that MMP1 expression is related with clinical prognosis and tumor immune infiltration, and MMP1 can become a prognostic and immunological biomarker.
Collapse
Affiliation(s)
- Shuai Mao
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Anliang Xia
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Xuewen Tao
- Department of Hepatobiliary Surgery, Medicine School of Southeast University Nanjing Drum Tower Hospital, Nanjing, China
| | - Dingde Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiamu Qu
- Department of Hepatobiliary Surgery, Medicine School of Southeast University Nanjing Drum Tower Hospital, Nanjing, China
| | - Meiling Sun
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Haowei Wei
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Guoqiang Li
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China,*Correspondence: Guoqiang Li,
| |
Collapse
|
10
|
ADAMTS1 as potential prognostic biomarker promotes malignant invasion of glioma. Int J Clin Oncol 2023; 28:52-68. [PMID: 36371587 DOI: 10.1007/s10147-022-02268-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Glioma is the most common intracranial malignancy in adults with a high degree of malignancy and poor prognosis, which is largely attributed to the existence of glioma stem cells (GSCs). Previous evidence indicated that the matrix metalloproteinase ADAMTS1 was implicated in the process of tumor invasion, but the involvement of ADAMTS1 in glioma malignant invasion remains poorly understood. METHODS The expression and prognosis values of ADAMTS1 were investigated in patients with glioma based on ONCOMINE and GEPIA databases. ADAMTS1 expression of different malignancy grade tissues was determined by immunohistochemistry. The effects of ADAMTS1 on cell proliferation and invasion were determined by clone formation assay and Transwell migration assay. The animal experiment was performed in an intracranial orthotopic xenograft model by knockout of ADAMTS1. Stemness properties and Notch1-SOX2 pathway were examined in stable ADAMTS1 knockdown GSCs. RESULTS The expression levels of ADAMTS1 were significantly higher in glioma tissues and significantly correlated with the grade of malignancy and prognosis of glioma. Elevated ADAMTS1 expression was associated with SOX2, N-cadherin and the resistance of chemoradiotherapy of glioma patients. ADAMTS1 knockout suppressed the intracranial orthotopic xenograft growth and prolonged the survival of xenograft mice in vivo. Mechanistically, we found a blockade of the migration and invasiveness of GSCs and the expression levels of Notch1 and SOX2 in absence of ADAMTS1. CONCLUSION As a biomarker for prediction of prognosis, ADAMTS1 may affect the invasive phenotype of GSCs by regulating Notch1-SOX2 signaling pathway, thereby promoting the invasive growth of glioma.
Collapse
|
11
|
Miyagi M, Katagiri H, Murata H, Wasa J, Takahashi T, Murakami H, Harada H, Mori K, Takahashi M. Osteosclerotic change as a therapeutic response to gefitinib in symptomatic non-small cell lung cancer bone metastasis. BMC Pulm Med 2022; 22:491. [PMID: 36581856 PMCID: PMC9801654 DOI: 10.1186/s12890-022-02226-1] [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: 04/11/2022] [Accepted: 11/05/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Despite improvement in the overall survival of patients with non-small cell lung cancer (NSCLC) harboring epidermal growth factor receptor (EGFR) mutation, the effects of EGFR tyrosine kinase inhibitor (EGFR-TKI) treatment on bone metastasis remain unclear. This study investigated radiological responses to gefitinib regarding bone metastasis in patients. METHODS We treated 260 patients with NSCLC and symptomatic bone metastasis. Thirty-seven patients harboring EGFR mutation were treated with gefitinib for more than 30 days and followed up for more than 3 months (GEF group). We performed a retrospective observational study by selecting 36 cases without EGFR-TKI treatment, at least 3 months of follow-up, and at least two radiological evaluations as the control group. We assessed the best overall radiological response, interval from treatment initiation to appearance of a radiological response, and the local response maintenance rate. RESULTS The best effect in the GEF group was 98% partial response or better, which was significantly higher than the 57% observed in the control group (p < 0.001). The GEF and control groups maintained 83% and 42% local response maintenance rates at one year, respectively (p < 0.001). In the GEF with radiotherapy group, the local response maintenance rate was maintained at 92% at 1 year, while in the GEF without RT group, there was a decrease in the local response maintenance rate from 270 days. CONCLUSION Gefitinib treatment for bone metastases in patients harboring EGFR mutation resulted in a beneficial osteosclerotic change in most patients. Combined gefitinib and radiotherapy provide long-lasting local control of bone metastases.
Collapse
Affiliation(s)
- Michihito Miyagi
- Devision of Orthopedic Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Hirohisa Katagiri
- Devision of Orthopedic Oncology, Shizuoka Cancer Center, Shizuoka, Japan.
| | - Hideki Murata
- Devision of Orthopedic Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Junji Wasa
- Devision of Orthopedic Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | | | - Haruyasu Murakami
- Devision of Thoracic Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Hideyuki Harada
- Division of Radiation Therapy, Radiation and Proton Therapy Center, Shizuoka Cancer Center, Shizuoka, Japan
| | - Keita Mori
- Department of Biostatistics, Clinical Research Center, Shizuoka Cancer Center, Shizuoka, Japan
| | - Mitsuru Takahashi
- Devision of Orthopedic Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
12
|
Mangiavini L, Peretti GM, Canciani B, Maffulli N. Epidermal growth factor signalling pathway in endochondral ossification: an evidence-based narrative review. Ann Med 2022; 54:37-50. [PMID: 34955078 PMCID: PMC8725985 DOI: 10.1080/07853890.2021.2015798] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
During endochondral bone development, a complex process that leads to the formation of the majority of skeletal elements, mesenchymal cells condense, differentiating into chondrocytes and producing the foetal growth plate. Chondrocytes progressively hypertrophy, induce angiogenesis and are then gradually replaced by bone. Epidermal Growth Factor (EGF), one of many growth factors, is the prototype of the EGF-ligand family, which comprises several proteins involved in cell proliferation, migration and survival. In bone, EGF pathway signalling finely tunes the first steps of chondrogenesis by maintaining mesenchymal cells in an undifferentiated stage, and by promoting hypertrophic cartilage replacement. Moreover, EGF signalling modulates bone homeostasis by stimulating osteoblast and osteoclast proliferation, and by regulating osteoblast differentiation under specific spatial and temporal conditions. This evidence-based narrative review describes the EGF pathway in bone metabolism and endochondral bone development. This comprehensive description may be useful in light of possible clinical applications in orthopaedic practice. A deeper knowledge of the role of EGF in bone may be useful in musculoskeletal conditions which may benefit from the modulation of this signalling pathway.Key messagesThe EGF pathway is involved in bone metabolism.EGF signalling is essential in the very early stages of limb development by maintaining cells in an undifferentiated stage.EGF pathway positively regulates chondrocyte proliferation, negatively modulates hypertrophy, and favours cartilage replacement by bone.EGF and EGF-like proteins finely tune the proliferation and differentiation of bone tissue cells, and they also regulate the initial phases of endochondral ossification.
Collapse
Affiliation(s)
- L Mangiavini
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical Sciences for Health, Università Degli Studi di Milano, Milan, Italy
| | - G M Peretti
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical Sciences for Health, Università Degli Studi di Milano, Milan, Italy
| | - B Canciani
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - N Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy.,Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Queen Mary University of London, London, UK.,School of Pharmacy and Bioengineering, Keele University Faculty of Medicine, Stoke on Trent, UK
| |
Collapse
|
13
|
Dai R, Liu M, Xiang X, Xi Z, Xu H. Osteoblasts and osteoclasts: an important switch of tumour cell dormancy during bone metastasis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:316. [PMID: 36307871 PMCID: PMC9615353 DOI: 10.1186/s13046-022-02520-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/18/2022] [Indexed: 11/30/2022]
Abstract
Bone metastasis occurs when tumour cells dissociate from primary tumours, enter the circulation (circulating tumour cells, CTCs), and colonize sites in bone (disseminated tumour cells, DTCs). The bone marrow seems to be a particularly dormancy-inducing environment for DTCs, yet the mechanisms of dormancy initiation, reactivation, and interaction within the bone marrow have to be elucidated. Intriguingly, some evidence has suggested that dormancy is a reversible state that is switched 'on' or 'off' depending on the presence of various bone marrow resident cells, particularly osteoclasts and osteoblasts. It has become clear that these two cells contribute to regulating dormant tumour cells in bone both directly (interaction) and indirectly (secreted factors). The involved mechanisms include TGFβ signalling, the Wnt signalling axis, the Notch2 pathway, etc. There is no detailed review that specifically focuses on ascertaining the dynamic interactions between tumour cell dormancy and bone remodelling. In addition, we highlighted the roles of inflammatory cytokines during this 'cell-to-cell' communication. We also discussed the potential clinical relevance of remodelling the bone marrow niche in controlling dormant tumour cells. Understanding the unique role of osteoclasts and osteoblasts in regulating tumour dormancy in bone marrow will provide new insight into preventing and treating tumour bone metastasis.
Collapse
Affiliation(s)
- Rongchen Dai
- grid.412540.60000 0001 2372 7462School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China ,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203 China
| | - Mengfan Liu
- grid.412540.60000 0001 2372 7462School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China ,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203 China
| | - Xincheng Xiang
- grid.47840.3f0000 0001 2181 7878Rausser College of Natural Resources, University of California Berkeley, Berkeley, CA 94720 USA
| | - Zhichao Xi
- grid.412540.60000 0001 2372 7462School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China ,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203 China
| | - Hongxi Xu
- grid.412585.f0000 0004 0604 8558Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| |
Collapse
|
14
|
Gene Expression and Immunochemistry Analysis of ADAMTS-1 and Versican in Ameloblastoma. Int J Dent 2022; 2022:5235376. [PMID: 36338393 PMCID: PMC9629950 DOI: 10.1155/2022/5235376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 09/23/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022] Open
Abstract
Background Ameloblastoma is a benign but locally invasive odontogenic epithelial tumor, associated with a high recurrence rate after treatment. The action of enzymes of the metalloproteinase family is important to the degraded extracellular matrix, contributing to invasion. Thus, this study aimed to investigate the gene and protein expression of ADAMTS-1 and versican in ameloblastoma. Materials and Methods Twenty cases of ameloblastoma (n = 20) and ten dental follicles (DF) (n = 10) were used as a source for immunochemistry and quantitative RT-PCR for determining the protein and mRNA expressions of the concerned genes, respectively. Moreover, western blot and indirect immunofluorescence analysis were performed in AME cells. Results ADAMTS-1 and versican were overexpressed in DF than ameloblastoma by RT-PCR. However, in the immunolocalization analysis, ADAMTS-1 was expressed in ameloblastoma more than in DF and versican immunostaining obtained a similar pattern between ameloblastoma and DF. Indirect immunofluorescence detected the ADAMTS-1 and versican expression in cell lines derived from ameloblastoma. Western blot from cell lysate and conditioned medium detected ADAMTS-1 bands representing full-length and different processed forms. Monensin treatment confined ADAMTS-1 in the cell cytoplasm. Versican fragments also were detected in different compartments, intracellular and conditioned medium, allowing the versican process by ADAMTS-1. Conclusion This study showed a distinct expression of ADAMTS-1 and versican in ameloblastoma and DF, with ADAMTS-1 protein higher expression observed in ameloblastoma and possibly cleaved versican. These findings suggested that ADAMTS-1 may participate in tumor invasion, especially for the degradation of substrates (versican) in the ECM.
Collapse
|
15
|
Zhou J, Wu L, Xu P, Li Y, Ji Z, Kang X. Filamin A Is a Potential Driver of Breast Cancer Metastasis via Regulation of MMP-1. Front Oncol 2022; 12:836126. [PMID: 35359350 PMCID: PMC8962737 DOI: 10.3389/fonc.2022.836126] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/14/2022] [Indexed: 01/01/2023] Open
Abstract
Recurrent metastasis is a major fatal cause of breast cancer. Regretfully, the driving force and the molecular beneath have not been fully illustrated yet. In this study, a cohort of breast cancer patients with locoregional metastasis was recruited. For them, we collected the matched samples of the primary tumor and metastatic tumor, and then we determined the mutation profiles with whole-exome sequencing (WES). On basis of the profiles, we identified a list of deleterious variants in eight susceptible genes. Of them, filamin A (FLNA) was considered a potential driver gene of metastasis, and its low expression could enhance 5 years’ relapse survival rate by 15%. To prove the finding, we constructed a stable FLNA knockout tumor cell line, which manifested that the cell abilities of proliferation, migration, and invasion were significantly weakened in response to the gene knockout. Subsequently, xenograft mouse experiments further proved that FLNA knockout could inhibit local or distal metastasis. Putting all the results together, we consolidated that FLNA could be a potential driver gene to metastasis of breast cancer, in particular triple-negative breast cancer. Additional experiments also suggested that FLNA might intervene in metastasis via the regulation of MMP-1 expression. In summary, this study demonstrates that FLNA may play as a positive regulator in cancer proliferation and recurrence. It provides new insight into breast cancer metastasis and suggests a potential new therapeutic target for breast cancer therapy.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lvying Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Pengyan Xu
- Department of Surgical Research, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Yue Li
- Department of Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhiliang Ji
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- *Correspondence: Xinmei Kang, ; Zhiliang Ji,
| | - Xinmei Kang
- Department of Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Xinmei Kang, ; Zhiliang Ji,
| |
Collapse
|
16
|
Schnellmann R. Advances in ADAMTS biomarkers. Adv Clin Chem 2022; 106:1-32. [PMID: 35152971 DOI: 10.1016/bs.acc.2021.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
A Disintegrin and Metalloproteinase with Thrombospondin motifs (ADAMTS) are major mediators in extracellular matrix (ECM) turnover and have gained increasing interest over the last years as major players in ECM remodeling during tissue homeostasis and the development of diseases. Although, ADAMTSs are recognized in playing important roles during tissue remodeling, and loss of function in various member of the ADAMTS family could be associated with the development of numerous diseases, limited knowledge is available about their specific substrates and mechanism of action. In this chapter, we will review current knowledge about ADAMTSs and their use as disease biomarkers.
Collapse
Affiliation(s)
- Rahel Schnellmann
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
17
|
Circulating cytokines present in multiple myeloma patients inhibit the osteoblastic differentiation of adipose stem cells. Leukemia 2022; 36:540-548. [PMID: 34556797 DOI: 10.1038/s41375-021-01428-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 09/02/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023]
Abstract
Myeloma is characterized by bone lesions, which are related to both an increased osteoclast activity and a defect in the differentiation of medullary mesenchymal stem cells (MSCs) into osteoblasts. Outside the medullary environment, adipocyte-derived MSCs (ASCs) could represent a source of functional osteoblasts. However, we recently found a defect in the osteoblastic differentiation of ASCs from myeloma patients (MM-ASCs). We examined the effects of plasma from myeloma patients at diagnosis (MM-plasmas) and in complete remission (CR-plasmas) and from healthy donors on the osteoblastic differentiation of healthy donor-derived ASCs (HD-ASCs). Osteoblastogenesis in HD-ASCs was suppressed by MM-plasmas. Seven cytokines (ANG1, ENA-78, EGF, PDGF-AA/AB/BB, and TARC) were increased in MM-plasmas and separately inhibited the osteoblastic differentiation of HD-ASCs. Comparison of MM-ASCs and HD-ASCs by RNA sequencing showed that two master genes characterizing adipocyte differentiation, CD36 and PPARγ, were upregulated in MM-ASCs as compared to HD-ASCs. Finally, we demonstrated a significant increase in CD36 and PPARγ expression in HD-ASCs in the presence of MM-plasmas or the seven cytokines individually, similarly as in MM-ASCs. We conclude that specific cytokines in MM-plasmas, besides the well-known DKK1, inhibit the osteoblastic differentiation of MM- and HD-ASCs with a skewing towards adipocyte differentiation.
Collapse
|
18
|
Cheng T, Chen P, Chen J, Deng Y, Huang C. Landscape Analysis of Matrix Metalloproteinases Unveils Key Prognostic Markers for Patients With Breast Cancer. Front Genet 2022; 12:809600. [PMID: 35069702 PMCID: PMC8770541 DOI: 10.3389/fgene.2021.809600] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/16/2021] [Indexed: 12/15/2022] Open
Abstract
Breast cancer (BRCA) is the most common cancer in the world, of which incidence rate and mortality are the highest in women. Being responsible for the remodeling and degradation of extracellular matrix proteins, matrix metalloproteinases (MMPs) have been regarded as one of the most important protease family related to tumorigenesis. It has been demonstrated that MMPs play crucial roles in some tumor invasion and metastasis. However, the potential roles of MMPs in tumorigenesis and progression of BRCA and its subtype remain elusive. Herein, we conducted a systematic study on MMPs via a series of database-based retrospective analysis, including TCGA, R Studio, GEPIA, Kaplan-Meier Plotter, cBioPortal, STRING, GeneMANIA and TIMER. As a result, many MMP family members were differentially expressed in patients with BRCA, e.g., the expressions of MMP1, MMP9, MMP11 and MMP13 were up-regulated, whereas the expression levels of MMP19 and MMP28 were down-regulated. MMP9, MMP12, MMP15 and MMP27 were significantly correlated with the clinical stages of BRCA, implying their important roles in the occurrence and development of BRCA. In addition, the survival analysis indicated that different expression pattern of MMPs exhibited distinct outcomes in patient with BRCA, e.g., patients with high expression of MMP2, MMP8, MMP16, MMP17, MMP19, MMP20, MMP21, MMP24, MMP25, MMP26 and MMP27 had a prolonged survival time, while the others (MMP1, MMP7, MMP9, MMP12 and MMP15) exhibited poor prognosis. Subsequent functional and network analysis revealed MMPs were mainly correlated with parathyroid hormone synthesis and secretion pathway, collagen metabolism, and their effect on the activities of serine hydrolase, serine peptidase and aminopeptidase. Notably, our analysis showed that the expression of MMPs was significantly correlated with the infiltration of various immune cells in BRCA, including CD8+T cells, CD4+T cells, macrophages, neutrophils, B cells, and dendritic cells, suggesting the close correlations between MMPs and immune functions. In short, our study disclosed MMPs play multiple biological roles in the development of BRCA, MMP1 and MMP9 might be used as independent prognostic markers and potential therapeutic targets for diagnosis and treatment for patients with BRCA.
Collapse
Affiliation(s)
- Tianyi Cheng
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Peiying Chen
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jingyi Chen
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Yingtong Deng
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Chen Huang
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China.,Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau, China
| |
Collapse
|
19
|
Coniglio SJ, Segall JE. Microglial-stimulation of glioma invasion involves the EGFR ligand amphiregulin. PLoS One 2021; 16:e0260252. [PMID: 34843542 PMCID: PMC8629255 DOI: 10.1371/journal.pone.0260252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 11/05/2021] [Indexed: 12/13/2022] Open
Abstract
High grade glioma is one of the deadliest human cancers with a median survival rate of only one year following diagnosis. The highly motile and invasive nature of high grade glioma makes it difficult to completely remove surgically. Therefore, increasing our knowledge of the mechanisms glioma cells use to invade normal brain is of critical importance in designing novel therapies. It was previously shown by our laboratory that tumor-associated microglia (TAMs) stimulate glioma cell invasion and this process is dependent on CSF-1R signaling. In this study, we seek to identify pro-invasive factors that are upregulated in microglia in a CSF-1R-dependent manner. We assayed cDNA and protein from microglia treated with conditioned media from the murine glioma cell line GL261, and discovered that several EGFR ligands including amphiregulin (AREG) are strongly upregulated. This upregulation is blocked by addition of a pharmacological CSF-1R inhibitor. Using RNA interference, we show that AREG-depleted microglia are less effective at promoting invasion of GL261 cells into Matrigel-coated invasion chambers. In addition, an AREG blocking antibody strongly attenuates the ability of THP-1 macrophages to activate human glioma cell line U87 invasion. Furthermore, we have identified a signaling pathway which involves CSF-1 signaling through ERK to upregulate AREG expression in microglia. Interfering with ERK using pharmacological inhibitors prevents AREG upregulation in microglia and microglia-stimulated GL261 invasion. These data highlight AREG as a key factor in produced by tumor associated microglia in promoting glioma invasion.
Collapse
Affiliation(s)
- Salvatore J. Coniglio
- New Jersey Center for Science Technology and Mathematics, Kean University, Union, NJ, United States of America
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Jeffrey E. Segall
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, United States of America
- Gruss Lipper Biophotonics Center, Bronx, NY, United States of America
| |
Collapse
|
20
|
Segura-Bautista D, Maya-Nunez G, Aguilar-Rojas A, Huerta-Reyes M, Pérez-Solis MA. Contribution of Stemness-linked Transcription Regulators to the Progression of Breast Cancer. Curr Mol Med 2021; 22:766-778. [PMID: 34819003 DOI: 10.2174/1566524021666211124154803] [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: 01/15/2021] [Revised: 05/05/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022]
Abstract
Although there are currently several factors that allow measuring the risk of having breast cancer or predicting its progression, the underlying causes of this malignancy have remained unknown. Several molecular studies have described some mechanisms involved in the progress of breast cancer. These have helped in identifying new targets with therapeutic potential. However, despite the therapeutic strategies implemented from the advances achieved in breast cancer research, a large percentage of patients with breast cancer die due to the spread of malignant cells to other tissues or organs, such as bones and lungs. Therefore, determining the processes that promote the migration of malignant cells remains one of the greatest challenges for oncological research. Several research groups have reported evidence on how the dedifferentiation of tumor cells leads to the acquisition of stemness characteristics, such as invasion, metastasis, the capability to evade the immunological response, and resistance to several cytotoxic drugs. These phenotypic changes have been associated with a complex reprogramming of gene expression in tumor cells during the Epithelial-Mesenchymal Transition (EMT). Considering the determining role that the transcriptional regulation plays in the expression of the specific characteristics and attributes of breast cancer during ETM, in the present work, we reviewed and analyzed several transcriptional mechanisms that support the mesenchymal phenotype. In the same way, we established the importance of transcription factors with a therapeutic perspective in the progress of breast cancer.
Collapse
Affiliation(s)
- David Segura-Bautista
- Medical Research Unit in Reproductive Medicine, UMAE Hospital de Gineco Obstetricia no. 4 'Luis Castelazo-Ayala', Instituto Mexicano del Seguro Social, Mexico City. Mexico
| | - Guadalupe Maya-Nunez
- Medical Research Unit in Reproductive Medicine, UMAE Hospital de Gineco Obstetricia no. 4 'Luis Castelazo-Ayala', Instituto Mexicano del Seguro Social, Mexico City. Mexico
| | - Arturo Aguilar-Rojas
- Medical Research Unit in Reproductive Medicine, UMAE Hospital de Gineco Obstetricia no. 4 'Luis Castelazo-Ayala', Instituto Mexicano del Seguro Social, Mexico City. Mexico
| | - Maira Huerta-Reyes
- Medical Research Unit in Nephrological Diseases, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City. Mexico
| | - Marco Allan Pérez-Solis
- Medical Research Unit in Reproductive Medicine, UMAE Hospital de Gineco Obstetricia no. 4 'Luis Castelazo-Ayala', Instituto Mexicano del Seguro Social, Mexico City. Mexico
| |
Collapse
|
21
|
Ban J, Fock V, Aryee DNT, Kovar H. Mechanisms, Diagnosis and Treatment of Bone Metastases. Cells 2021; 10:2944. [PMID: 34831167 PMCID: PMC8616226 DOI: 10.3390/cells10112944] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022] Open
Abstract
Bone and bone marrow are among the most frequent metastatic sites of cancer. The occurrence of bone metastasis is frequently associated with a dismal disease outcome. The prevention and therapy of bone metastases is a priority in the treatment of cancer patients. However, current therapeutic options for patients with bone metastatic disease are limited in efficacy and associated with increased morbidity. Therefore, most current therapies are mainly palliative in nature. A better understanding of the underlying molecular pathways of the bone metastatic process is warranted to develop novel, well-tolerated and more successful treatments for a significant improvement of patients' quality of life and disease outcome. In this review, we provide comparative mechanistic insights into the bone metastatic process of various solid tumors, including pediatric cancers. We also highlight current and innovative approaches to biologically targeted therapy and immunotherapy. In particular, we discuss the role of the bone marrow microenvironment in the attraction, homing, dormancy and outgrowth of metastatic tumor cells and the ensuing therapeutic implications. Multiple signaling pathways have been described to contribute to metastatic spread to the bone of specific cancer entities, with most knowledge derived from the study of breast and prostate cancer. However, it is likely that similar mechanisms are involved in different types of cancer, including multiple myeloma, primary bone sarcomas and neuroblastoma. The metastatic rate-limiting interaction of tumor cells with the various cellular and noncellular components of the bone-marrow niche provides attractive therapeutic targets, which are already partially exploited by novel promising immunotherapies.
Collapse
Affiliation(s)
- Jozef Ban
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
| | - Valerie Fock
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
| | - Dave N. T. Aryee
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
- Department of Pediatrics, Medical University Vienna, 1090 Vienna, Austria
| | - Heinrich Kovar
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
- Department of Pediatrics, Medical University Vienna, 1090 Vienna, Austria
| |
Collapse
|
22
|
Zuo H, Yang D, Wan Y. Fam20C Regulates Bone Resorption and Breast Cancer Bone Metastasis through Osteopontin and BMP4. Cancer Res 2021; 81:5242-5254. [PMID: 34433585 DOI: 10.1158/0008-5472.can-20-3328] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/14/2020] [Accepted: 08/23/2021] [Indexed: 11/16/2022]
Abstract
Fam20C is a kinase that generates the majority of secreted phosphoproteins and regulates biomineralization. However, its potential roles in bone resorption and breast cancer bone metastasis are unknown. Here we show that Fam20C in the myeloid lineage suppresses osteoclastogenesis and bone resorption, during which, osteopontin (OPN) is the most abundant phosphoprotein secreted in a Fam20C-dependent manner. OPN phosphorylation by Fam20C decreased OPN secretion, and OPN neutralization reduced Fam20C deficiency-induced osteoclast differentiation and bone metastasis. In contrast, Fam20C in breast cancer cells promoted bone metastasis by facilitating the phosphorylation and secretion of BMP4, which in turn enhanced osteoclastogenesis. Mutation of the BMP4 phosphorylation site elevated BMP4 lysosomal degradation and reduced BMP4 secretion. In breast cancer cells, BMP4 depletion or treatment with a BMP4 signaling inhibitor diminished osteoclast differentiation and bone metastasis and abolished Fam20C-mediated regulation of these processes. Collectively, this study discovers distinct roles for Fam20C in myeloid cells and breast cancer cells and highlights OPN and BMP4 as potential therapeutic targets for breast cancer bone metastasis. SIGNIFICANCE: Osteoclastogenesis and bone metastasis are suppressed by myeloid-derived Fam20C, but enhanced by breast cancer-associated Fam20C, uncovering novel Fam20C functions and new therapeutic strategies via targeting Fam20C substrates OPN and BMP4.
Collapse
Affiliation(s)
- Hao Zuo
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Dengbao Yang
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yihong Wan
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
23
|
Kapoor R, Saxena AK, Vasudev P, Sundriyal D, Kumar A. Cancer induced bone pain: current management and future perspectives. Med Oncol 2021; 38:134. [PMID: 34581894 DOI: 10.1007/s12032-021-01587-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/18/2021] [Indexed: 12/31/2022]
Abstract
Recent improvements in the therapeutic armamentarium of oncology by the addition of targeted and immunotherapeutic agents have led to an increase in the life expectancy of advanced-stage cancer patients. This has led to an increased number of patients presenting with bone metastasis and experiencing episodes of cancer-induced bone pain (CIBP). CIBP is a crippling, chronic, morbid state interfering significantly with the functional capacity and the quality of life (QoL). CIBP is characterized by a complex multifactorial pathophysiological mechanism involving tumor cells, bone cells, inflammatory microenvironment, and the neuronal tissue. It may not be possible to mitigate pain completely; therefore, the aim should be to reach the lowest possible level of pain that allows for an acceptable QoL to the patient. Multimodality approach of surgical, radiation, medical and behavioral techniques is thus recommended to manage CIBP. This review discusses the pathogenesis and pathophysiological mechanism accompanying bone metastasis and CIBP, currently approved therapies for the management of CIBP, and the future perspective.
Collapse
Affiliation(s)
- Ruchi Kapoor
- Department of Anesthesiology and Pain Medicine, University College of Medical Sciences and GTB Hospital, New Delhi, India
| | - Ashok Kumar Saxena
- Department of Anesthesiology and Pain Medicine, University College of Medical Sciences and GTB Hospital, New Delhi, India
| | - Prerna Vasudev
- Department of Anesthesiology and Pain Medicine, University College of Medical Sciences and GTB Hospital, New Delhi, India
| | - Deepak Sundriyal
- Department of Medical Oncology, Hematology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India.
| | - Arvind Kumar
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| |
Collapse
|
24
|
Zhang HJ, Liao HY, Bai DY, Wang ZQ, Xie XW. MAPK /ERK signaling pathway: A potential target for the treatment of intervertebral disc degeneration. Biomed Pharmacother 2021; 143:112170. [PMID: 34536759 DOI: 10.1016/j.biopha.2021.112170] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/01/2021] [Accepted: 09/05/2021] [Indexed: 12/17/2022] Open
Abstract
Intervertebral disc degeneration (IDD) is a chronic skeletal muscle degenerative disease, which is considered the main cause of low back pain. It seriously affects the quality of life of patients and consequently brings a heavy economic burden to their families and the society. Although IDD is considered a natural process in degenerative lesions, it is mainly caused by aging, trauma, genetic susceptibility and other factors. It is closely related to changes in the tissue structure and function, including the progressive destruction of extracellular matrix, cell aging, cell death of the intervertebral disc (IVD), inflammation, and impairment of tissue biomechanical function. Currently, the treatment of IDD is aimed at alleviating symptoms rather than at targeting pathological changes in the IVD. Furthermore, the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathway is closely related to various pathological processes in IDD, and the activation of the MAPK/ERK pathway promotes the degradation of the IVD extracellular matrix, cell aging, apoptosis, and inflammatory responses. It also induces autophagy and oxidative stress that accelerate the IVD process. In our current review, we summarize the latest developments in the negative regulation of IDD after activation of the MAPK/ERK signaling pathway and emphasize on its influence on IDD. Targeting this pathway may become an attractive treatment strategy for IDD in the near future.
Collapse
Affiliation(s)
- Hai-Jun Zhang
- Second Provincial People's Hospital of Gansu, 1 Hezheng West Street, Lanzhou 730000, PR China; Affiliated Hospital of Northwest Minzu Univsity, Lanzhou 730000, PR China
| | - Hai-Yang Liao
- Fist Affiliated Hospital of Ganan Medical University, 23 Qingnian Road, Ganzhou 342800, PR China
| | - Deng-Yan Bai
- Second Provincial People's Hospital of Gansu, 1 Hezheng West Street, Lanzhou 730000, PR China; Affiliated Hospital of Northwest Minzu Univsity, Lanzhou 730000, PR China
| | - Zhi-Qiang Wang
- Fist Affiliated Hospital of Ganan Medical University, 23 Qingnian Road, Ganzhou 342800, PR China
| | - Xing-Wen Xie
- Second Provincial People's Hospital of Gansu, 1 Hezheng West Street, Lanzhou 730000, PR China; Affiliated Hospital of Northwest Minzu Univsity, Lanzhou 730000, PR China.
| |
Collapse
|
25
|
Cheng X, Wei J, Ge Q, Xing D, Zhou X, Qian Y, Jiang G. The optimized drug delivery systems of treating cancer bone metastatic osteolysis with nanomaterials. Drug Deliv 2021; 28:37-53. [PMID: 33336610 PMCID: PMC7751395 DOI: 10.1080/10717544.2020.1856225] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Some cancers such as human breast cancer, prostate cancer, and lung cancer easily metastasize to bone, leading to osteolysis and bone destruction accompanied by a complicated microenvironment. Systemic administration of bisphosphonates (BP) or denosumab is the routine therapy for osteolysis but with non-negligible side effects such as mandibular osteonecrosis and hypocalcemia. Thus, it is imperative to exploit optimized drug delivery systems, and some novel nanotechnology and nanomaterials have opened new horizons for scientists. Targeted and local drug delivery systems can optimize biodistribution depending on nanoparticles (NPs) or microspheres (MS) and implantable biomaterials with the controllable property. Drug delivery kinetics can be optimized by smart and sustained/local drug delivery systems for responsive delivery and sustained delivery. These delicately fabricated drug delivery systems with special matrix, structure, morphology, and modification can minimize unexpected toxicity caused by systemic delivery and achieve desired effects through integrating multiple drugs or multiple functions. This review summarized recent studies about optimized drug delivery systems for the treatment of cancer metastatic osteolysis, aimed at giving some inspiration in designing efficient multifunctional drug delivery systems.
Collapse
Affiliation(s)
- Xi Cheng
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jinrong Wei
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Qi Ge
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Danlei Xing
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xuefeng Zhou
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, People's Republic of China
| | - Yunzhu Qian
- Center of Stomatology, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Guoqin Jiang
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
26
|
Deasy SK, Erez N. A glitch in the matrix: organ-specific matrisomes in metastatic niches. Trends Cell Biol 2021; 32:110-123. [PMID: 34479765 DOI: 10.1016/j.tcb.2021.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023]
Abstract
Modification of the extracellular matrix (ECM) is a critical aspect of developing a metastasis-supportive organ niche. Recent work investigating ECM changes that facilitate metastasis has revealed ways in which different metastatic organ niches are similar as well as the distinct characteristics that make them unique. In this review, we present recent findings regarding how ECM modifications support metastasis in four frequent metastatic sites: the lung, liver, bone, and brain. We discuss ways in which these modifications are shared between metastatic organs as well as features specific to each location. We also discuss areas of technical innovation that could be advantageous to future research and areas of inquiry that merit further investigation.
Collapse
Affiliation(s)
- Sarah K Deasy
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
27
|
Othman A, Winogradzki M, Lee L, Tandon M, Blank A, Pratap J. Bone Metastatic Breast Cancer: Advances in Cell Signaling and Autophagy Related Mechanisms. Cancers (Basel) 2021; 13:cancers13174310. [PMID: 34503118 PMCID: PMC8431094 DOI: 10.3390/cancers13174310] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022] Open
Abstract
Bone metastasis is a frequent complication of breast cancer with nearly 70% of metastatic breast cancer patients developing bone metastasis during the course of their disease. The bone represents a dynamic microenvironment which provides a fertile soil for disseminated tumor cells, however, the mechanisms which regulate the interactions between a metastatic tumor and the bone microenvironment remain poorly understood. Recent studies indicate that during the metastatic process a bidirectional relationship between metastatic tumor cells and the bone microenvironment begins to develop. Metastatic cells display aberrant expression of genes typically reserved for skeletal development and alter the activity of resident cells within the bone microenvironment to promote tumor development, resulting in the severe bone loss. While transcriptional regulation of the metastatic process has been well established, recent findings from our and other research groups highlight the role of the autophagy and secretory pathways in interactions between resident and tumor cells during bone metastatic tumor growth. These reports show high levels of autophagy-related markers, regulatory factors of the autophagy pathway, and autophagy-mediated secretion of matrix metalloproteinases (MMP's), receptor activator of nuclear factor kappa B ligand (RANKL), parathyroid hormone related protein (PTHrP), as well as WNT5A in bone metastatic breast cancer cells. In this review, we discuss the recently elucidated mechanisms and their crosstalk with signaling pathways, and potential therapeutic targets for bone metastatic disease.
Collapse
|
28
|
The Roadmap of RANKL/RANK Pathway in Cancer. Cells 2021; 10:cells10081978. [PMID: 34440747 PMCID: PMC8393235 DOI: 10.3390/cells10081978] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 01/02/2023] Open
Abstract
The receptor activator of the nuclear factor-κB ligand (RANKL)/RANK signaling pathway was identified in the late 1990s and is the key mediator of bone remodeling. Targeting RANKL with the antibody denosumab is part of the standard of care for bone loss diseases, including bone metastases (BM). Over the last decade, evidence has implicated RANKL/RANK pathway in hormone and HER2-driven breast carcinogenesis and in the acquisition of molecular and phenotypic traits associated with breast cancer (BCa) aggressiveness and poor prognosis. This marked a new era in the research of the therapeutic use of RANKL inhibition in BCa. RANKL/RANK pathway is also an important immune mediator, with anti-RANKL therapy recently linked to improved response to immunotherapy in melanoma, non-small cell lung cancer (NSCLC), and renal cell carcinoma (RCC). This review summarizes and discusses the pre-clinical and clinical evidence of the relevance of the RANKL/RANK pathway in cancer biology and therapeutics, focusing on bone metastatic disease, BCa onset and progression, and immune modulation.
Collapse
|
29
|
Kim YJ. Immunomodulatory Effects of Human Colostrum and Milk. Pediatr Gastroenterol Hepatol Nutr 2021; 24:337-345. [PMID: 34316468 PMCID: PMC8279828 DOI: 10.5223/pghn.2021.24.4.337] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/27/2021] [Accepted: 05/17/2021] [Indexed: 01/03/2023] Open
Abstract
The immune system is not fully developed in human neonates and infants; breastfeeding is important in this stage as the bioactive components of human breast milk are known to have anti-microbial, anti-inflammatory, and immunomodulatory effects, and can therefore contribute to an infant's immunity against allergies, asthma, autoimmune diseases, and inflammatory bowel disease. Herein, the positive effect on the immune system by human colostrum and milk are reviewed.
Collapse
Affiliation(s)
- Yong Joo Kim
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
30
|
DNMT3A-mediated silence in ADAMTS9 expression is restored by RNF180 to inhibit viability and motility in gastric cancer cells. Cell Death Dis 2021; 12:428. [PMID: 33931579 PMCID: PMC8087691 DOI: 10.1038/s41419-021-03628-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 12/24/2022]
Abstract
ADAMTS9 belongs to the ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) protein family, and its expression is frequently silenced due to promoter hypermethylation in various human cancers. However, the underlying mechanisms remain largely unknown. In this study, we investigated the inhibitory effects of ADAMTS9 on gastric cancer (GC) cells. We initially examined ADAMTS9 protein level in 135 GC and adjacent normal tissue pairs, showing that ADAMTS9 was strikingly decreased in the malignant specimens and patients with low ADAMTS9 expression exhibited more malignant phenotypes and poorer outcome. ADAMTS9 expression was restored in AGS and BGC-823 cells, which then markedly suppressed cellular viability and motility in vitro and in vivo. As ADAMTS9 was enriched in the nuclei of gastric mucosal cells, RNA-sequencing experiment showed that ADAMTS9 significantly altered gene expression profile in BGC-823 cells. Additionally, DNA methyltransferase 3α (DNMT3A) was identified to be responsible for the hypermethylation of ADAMTS9 promoter, and this methyltransferase was ubiquitinated by ring finger protein 180 (RNF180) and then subject to proteasome-mediated degradation. In conclusion, we uncovered RNF180/DNMT3A/ADAMTS9 axis in GC cells and showed how the signaling pathway affected GC cells.
Collapse
|
31
|
Patil KC, Soekmadji C. Extracellular Vesicle-Mediated Bone Remodeling and Bone Metastasis: Implications in Prostate Cancer. Subcell Biochem 2021; 97:297-361. [PMID: 33779922 DOI: 10.1007/978-3-030-67171-6_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone metastasis is the tendency of certain primary tumors to spawn and dictate secondary neoplasia in the bone. The process of bone metastasis is regulated by the dynamic crosstalk between metastatic cancer cells, cellular components of the bone marrow microenvironment (osteoblasts, osteoclasts, and osteocytes), and the bone matrix. The feed-forward loop mechanisms governs the co-option of homeostatic bone remodeling by cancer cells in bone. Recent developments have highlighted the discovery of extracellular vesicles (EVs) and their diverse roles in distant outgrowths. Several studies have implicated EV-mediated interactions between cancer cells and the bone microenvironment in synergistically promoting pathological skeletal metabolism in the metastatic site. Nevertheless, the potential role that EVs serve in arbitrating intricate sequences of coordinated events within the bone microenvironment remains an emerging field. In this chapter, we review the role of cellular participants and molecular mechanisms in regulating normal bone physiology and explore the progress of current research into bone-derived EVs in directly triggering and coordinating the processes of physiological bone remodeling. In view of the emerging role of EVs in interorgan crosstalk, this review also highlights the multiple systemic pathophysiological processes orchestrated by the EVs to direct organotropism in bone in prostate cancer. Given the deleterious consequences of bone metastasis and its clinical importance, in-depth knowledge of the multifarious role of EVs in distant organ metastasis is expected to open new possibilities for prognostic evaluation and therapeutic intervention for advanced bone metastatic prostate cancer.
Collapse
Affiliation(s)
- Kalyani C Patil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Carolina Soekmadji
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia. .,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
32
|
Kim YJ, Oremus M, Chen HH, McFarlane T, Fearon D, Horton S. Factors affecting treatment selection and overall survival for first-line EGFR-tyrosine kinase inhibitor therapy in non-small-cell lung cancer. J Comp Eff Res 2021; 10:193-206. [PMID: 33543641 DOI: 10.2217/cer-2020-0173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: To investigate the factors associated with treatment selection and overall survival for first-line EGFR-tyrosine kinase inhibitors (EGFR-TKIs) therapy among patients with non-small-cell lung cancer. Materials & methods: We conducted a retrospective cohort study of linked administrative health databases in Ontario, Canada. Results: A total of 1011 patients received an EGFR-TKI as first-line therapy. Treatment selection and overall survival associated with these treatments were affected by age, sex, geographical residency, comorbidities and different sites of metastasis. Conclusion: Though recent approval of osimertinib offers a potential new standard of care in the first-line setting, earlier generation TKIs remain pillars in treatment of non-small-cell lung cancer therapeutic armamentarium. Our findings may contribute to optimizing treatment sequencing of EGFR-TKIs to maximize clinical benefits.
Collapse
Affiliation(s)
- Yong-Jin Kim
- School of Public Health & Health Systems, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Mark Oremus
- School of Public Health & Health Systems, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Helen H Chen
- School of Public Health & Health Systems, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Thomas McFarlane
- School of Pharmacy, University of Waterloo, Kitchener, ON N2G 1C5, Canada
| | - Danielle Fearon
- School of Public Health & Health Systems, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Susan Horton
- School of Public Health & Health Systems, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
33
|
Abstract
Breast cancer is the most common malignancy in women. Basic and translational breast cancer research relies heavily on experimental animal models. Ideally, such models for breast cancer should have commonality with human breast cancer in terms of tumor etiology, biological behavior, pathology, and response to therapeutics. This review introduces current progress in different breast cancer experimental animal models and analyzes their characteristics, advantages, disadvantages, and potential applications. Finally, we propose future research directions for breast cancer animal models.
Collapse
Affiliation(s)
- Li Zeng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Wei Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Ce-Shi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China. E-mail:
| |
Collapse
|
34
|
Shimoda M, Ohtsuka T, Okada Y, Kanai Y. Stromal metalloproteinases: Crucial contributors to the tumor microenvironment. Pathol Int 2020; 71:1-14. [PMID: 33074556 DOI: 10.1111/pin.13033] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/25/2020] [Indexed: 12/30/2022]
Abstract
Proteolytic balance is crucial for the maintenance of tissue homeostasis. In cancer, dysregulated proteolysis is involved in unregulated tissue remodeling and inflammation, leading to the promotion of tumor growth, local invasion, and metastasis. Metalloproteinases, which were first identified as collagen cleaving enzymes, have been shown to extensively degrade extracellular matrix proteins or selectively release cell surface-bound cytokines, growth factors, or their receptors, thereby impacting extracellular matrix integrity, immune cell recruitment and tissue turnover. Although tumor cells produce various metalloproteinases, the major source is thought to be stromal cells infiltrating the tumor. Different types of stromal cells express specific sets of metalloproteinases and their inhibitors, which specifically alter the milieu within the tumor. In this review, recent findings and knowledge regarding metalloproteinases derived from stromal cells during the creation of the tumor microenvironment are described and their contribution to the tumor progression and metastasis discussed.
Collapse
Affiliation(s)
- Masayuki Shimoda
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Ohtsuka
- Division of Thoracic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasunori Okada
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
35
|
Winkler J, Abisoye-Ogunniyan A, Metcalf KJ, Werb Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat Commun 2020; 11:5120. [PMID: 33037194 PMCID: PMC7547708 DOI: 10.1038/s41467-020-18794-x] [Citation(s) in RCA: 1021] [Impact Index Per Article: 255.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
Tissues are dynamically shaped by bidirectional communication between resident cells and the extracellular matrix (ECM) through cell-matrix interactions and ECM remodelling. Tumours leverage ECM remodelling to create a microenvironment that promotes tumourigenesis and metastasis. In this review, we focus on how tumour and tumour-associated stromal cells deposit, biochemically and biophysically modify, and degrade tumour-associated ECM. These tumour-driven changes support tumour growth, increase migration of tumour cells, and remodel the ECM in distant organs to allow for metastatic progression. A better understanding of the underlying mechanisms of tumourigenic ECM remodelling is crucial for developing therapeutic treatments for patients. Tumors are more than cancer cells — the extracellular matrix is a protein structure that organizes all tissues and is altered in cancer. Here, the authors review recent progress in understanding how the cancer cells and tumor-associated stroma cells remodel the extracellular matrix to drive tumor growth and metastasis.
Collapse
Affiliation(s)
- Juliane Winkler
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA.
| | - Abisola Abisoye-Ogunniyan
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA
| | - Kevin J Metcalf
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA
| | - Zena Werb
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA
| |
Collapse
|
36
|
Wen YC, Lin YW, Chu CY, Yang YC, Yang SF, Liu YF, Hsiao M, Lee WJ, Chien MH. Melatonin-triggered post-transcriptional and post-translational modifications of ADAMTS1 coordinately retard tumorigenesis and metastasis of renal cell carcinoma. J Pineal Res 2020; 69:e12668. [PMID: 32408377 DOI: 10.1111/jpi.12668] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/25/2020] [Accepted: 05/07/2020] [Indexed: 12/27/2022]
Abstract
A disintegrin and metalloprotease with thrombospondin motifs (ADAMTS) family are widely implicated in tissue remodeling events manifested in cancer development. ADAMTS1, the most fully characterized ADAMTS, plays conflicting roles in different cancer types; however, the role of ADAMTS1 in renal cell carcinoma (RCC) remains unclear. Herein, we found that ADAMTS1 is highly expressed in RCC tissues compared to normal renal tissues, and its expression was correlated with an advanced stage and a poor prognosis of RCC patients. In vitro, we observed higher expression of ADAMTS1 in metastatic (m)RCC cells compared to primary cells, and manipulation of ADAMTS1 expression affected cell invasion and clonogenicity. Results from protease array showed that ADAMTS1 is modulated by melatonin through mechanisms independent of the MT1 receptor in mRCC cells, and overexpression of ADAMTS1 relieved the invasion/clonogenicity and growth/metastasis inhibition imposed by melatonin treatment in vitro and in an orthotopic xenograft model. The human microRNA (miR) OneArray showed that miR-181d and miR-let-7f were induced by melatonin and, respectively, targeted the 3'-UTR and non-3'-UTR of ADAMTS1 to suppress its expression and mRCC invasive ability. Clinically, RCC patients with high levels of miR-181d or miR-let-7f and a low level of ADAMTS1 had the most favorable prognoses. In addition, ubiquitin/proteasome-mediated degradation of ADAMTS1 can also be triggered by melatonin. Together, our study indicates that ADAMTS1 may be a useful biomarker for predicting RCC progression. The novel convergence between melatonin and ADAMTS1 post-transcriptional and post-translational regulation provides new insights into the role of melatonin-induced molecular regulation in suppressing RCC progression.
Collapse
Affiliation(s)
- Yu-Ching Wen
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
| | - Yung-Wei Lin
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
| | - Chih-Ying Chu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chieh Yang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yu-Fan Liu
- Department of Biomedical Sciences, College of Medicine Sciences and Technology, Chung Shan Medical University, Taichung, Taiwan
- Division of Allergy, Department of Pediatrics, Chung-Shan Medical University Hospital, Taichung, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Jiunn Lee
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital Taipei, Taiwan
| |
Collapse
|
37
|
Monteran L, Ershaid N, Sabah I, Fahoum I, Zait Y, Shani O, Cohen N, Eldar-Boock A, Satchi-Fainaro R, Erez N. Bone metastasis is associated with acquisition of mesenchymal phenotype and immune suppression in a model of spontaneous breast cancer metastasis. Sci Rep 2020; 10:13838. [PMID: 32796899 PMCID: PMC7429866 DOI: 10.1038/s41598-020-70788-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023] Open
Abstract
The most common site of breast cancer metastasis is the bone, occurring in approximately 70% of patients with advanced disease. Bone metastasis is associated with severe morbidities and high mortality. Therefore, deeper understanding of the mechanisms that enable bone-metastatic relapse are urgently needed. We report the establishment and characterization of a bone-seeking variant of breast cancer cells that spontaneously forms aggressive bone metastases following surgical resection of primary tumor. We characterized the modifications in the immune milieu during early and late stages of metastatic relapse and found that the formation of bone metastases is associated with systemic changes, as well as modifications of the bone microenvironment towards an immune suppressive milieu. Furthermore, we characterized the intrinsic changes in breast cancer cells that facilitate bone-tropism and found that they acquire mesenchymal and osteomimetic features. This model provides a clinically relevant platform to study the functional interactions between breast cancer cells and the bone microenvironment, in an effort to identify novel targets for intervention.
Collapse
Affiliation(s)
- Lea Monteran
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Nour Ershaid
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Idan Sabah
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Ibrahim Fahoum
- Department of Pathology, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Yael Zait
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Ophir Shani
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Noam Cohen
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Anat Eldar-Boock
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Neta Erez
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel.
| |
Collapse
|
38
|
Kovar H, Bierbaumer L, Radic-Sarikas B. The YAP/TAZ Pathway in Osteogenesis and Bone Sarcoma Pathogenesis. Cells 2020; 9:E972. [PMID: 32326412 PMCID: PMC7227004 DOI: 10.3390/cells9040972] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/10/2020] [Accepted: 04/11/2020] [Indexed: 12/14/2022] Open
Abstract
YAP and TAZ are intracellular messengers communicating multiple interacting extracellular biophysical and biochemical cues to the transcription apparatus in the nucleus and back to the cell/tissue microenvironment interface through the regulation of cytoskeletal and extracellular matrix components. Their activity is negatively and positively controlled by multiple phosphorylation events. Phenotypically, they serve an important role in cellular plasticity and lineage determination during development. As they regulate self-renewal, proliferation, migration, invasion and differentiation of stem cells, perturbed expression of YAP/TAZ signaling components play important roles in tumorigenesis and metastasis. Despite their high structural similarity, YAP and TAZ are functionally not identical and may play distinct cell type and differentiation stage-specific roles mediated by a diversity of downstream effectors and upstream regulatory molecules. However, YAP and TAZ are frequently looked at as functionally redundant and are not sufficiently discriminated in the scientific literature. As the extracellular matrix composition and mechanosignaling are of particular relevance in bone formation during embryogenesis, post-natal bone elongation and bone regeneration, YAP/TAZ are believed to have critical functions in these processes. Depending on the differentiation stage of mesenchymal stem cells during endochondral bone development, YAP and TAZ serve distinct roles, which are also reflected in bone tumors arising from the mesenchymal lineage at different developmental stages. Efforts to clinically translate the wealth of available knowledge of the pathway for cancer diagnostic and therapeutic purposes focus mainly on YAP and TAZ expression and their role as transcriptional co-activators of TEAD transcription factors but rarely consider the expression and activity of pathway modulatory components and other transcriptional partners of YAP and TAZ. As there is a growing body of evidence for YAP and TAZ as potential therapeutic targets in several cancers, we here interrogate the applicability of this concept to bone tumors. To this end, this review aims to summarize our current knowledge of YAP and TAZ in cell plasticity, normal bone development and bone cancer.
Collapse
Affiliation(s)
- Heinrich Kovar
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (L.B.); (B.R.-S.)
- Department of Pediatrics, Medical University Vienna, 1090 Vienna, Austria
| | - Lisa Bierbaumer
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (L.B.); (B.R.-S.)
| | - Branka Radic-Sarikas
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (L.B.); (B.R.-S.)
| |
Collapse
|
39
|
Zuo H, Yang D, Yang Q, Tang H, Fu YX, Wan Y. Differential regulation of breast cancer bone metastasis by PARP1 and PARP2. Nat Commun 2020; 11:1578. [PMID: 32221289 PMCID: PMC7101362 DOI: 10.1038/s41467-020-15429-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/11/2020] [Indexed: 11/25/2022] Open
Abstract
PARP1 and PARP2 dual inhibitors, such as olaparib, have been recently FDA approved for the treatment of advanced breast and ovarian cancers. However, their effects on bone mass and bone metastasis are unknown. Here we show that olaparib increases breast cancer bone metastasis through PARP2, but not PARP1, specifically in the myeloid lineage, but not in the cancer cells. Olaparib treatment or PARP1/2 deletion promotes osteoclast differentiation and bone loss. Intriguingly, myeloid deletion of PARP2, but not PARP1, increases the population of immature myeloid cells in bone marrow, and impairs the expression of chemokines such as CCL3 through enhancing the transcriptional repression by β-catenin. Compromised CCL3 production in turn creates an immune-suppressive milieu by altering T cell subpopulations. Our findings warrant careful examination of current PARP inhibitors on bone metastasis and bone loss, and suggest cotreatment with CCL3, β-catenin inhibitors, anti-RANKL or bisphosphonates as potential combination therapy for PARP inhibitors.
Collapse
Affiliation(s)
- Hao Zuo
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Dengbao Yang
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Qiwen Yang
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Haidong Tang
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yang-Xin Fu
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yihong Wan
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Simmons Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
40
|
Zhou KQ, Liu WF, Yang LX, Sun YF, Hu J, Chen FY, Zhou C, Zhang XY, Peng YF, Yu L, Zhou J, Fan J, Wang Z. Circulating osteopontin per tumor volume as a prognostic biomarker for resectable intrahepatic cholangiocarcinoma. Hepatobiliary Surg Nutr 2019; 8:582-596. [PMID: 31929985 DOI: 10.21037/hbsn.2019.03.14] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background The role of osteopontin (OPN) in intrahepatic cholangiocarcinoma (ICC) remains controversial. This study aimed to explore the prognostic value of OPN in patients with ICC undergoing curative resection. Methods Patients undergoing curative resection from 2005 to 2016 were identified for inclusion in this retrospective study. The expression level of OPN in tumors was measured in each of the 228 patients by immunohistochemistry. Circulating OPN in serum was tested in 124 patients by ELISA. Tumor volume was calculated according to preoperative imaging or operation record. Proliferation assay, wound healing assay, and invasion assay were performed to investigate the biological function. Results Low expression of OPN in tissue was associated with lymph node metastasis (P=0.009) and shorter overall survival (OS) (P=0.001). A low level of circulating OPN/volume was associated with multiple tumors (P<0.001), vascular invasion (P=0.027), visceral peritoneal perforation (P=0.001), and lymph node metastasis (P=0.002). It was also able to predict the invasive behavior, lymph node metastasis, and early recurrence with the area under the receiver operating curve (AUC) of being 0.719, 0.708 and 0.622 respectively. Patients with a low level of circulating OPN/volume had shorter OS (P=0.028) and disease-free survival (DFS) (P=0.004) and could benefit from adjuvant chemotherapy (P=0.011). Compared with negative controlled cells, ICC cell lines, which expressed more OPN, showed a decelerated proliferation rate, the weaker ability of migration and invasion, while the opposite was true for the cells expressed less OPN. MMP1, MMP10, and CXCR4 were negatively regulated by OPN. Conclusions A low level of circulating OPN/volume could indicate aggressive characteristics, along with poor prognosis and efficacy of adjuvant chemotherapy in ICC patients. Over expression of OPN may inhibit phenotypes facilitating ICC metastasis by negatively regulating MMP1, MMP10, and CXCR4.
Collapse
Affiliation(s)
- Kai-Qian Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China
| | - Wei-Feng Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China.,Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Liu-Xiao Yang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China
| | - Yun-Fan Sun
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China.,Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jie Hu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China.,Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Fei-Yu Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China
| | - Cheng Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China
| | - Xiang-Yu Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China
| | - Yuan-Fei Peng
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China.,Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lei Yu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China.,Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jian Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China.,Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Jia Fan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China.,Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Zheng Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, China.,Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
41
|
Han Z, Zhan R, Chen S, Deng J, Shi J, Wang W. miR-181b/Oncostatin m axis inhibits prostate cancer bone metastasis via modulating osteoclast differentiation. J Cell Biochem 2019; 121:1664-1674. [PMID: 31680294 DOI: 10.1002/jcb.29401] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022]
Abstract
The activation of osteoblasts is significantly correlated to prostate tumor bone metastasis and bone loss. Oncostatin M (OSM) could promote breast cancer metastasis to bone. However, its role and mechanism in prostate cancer bone metastasis remain unclear. MicroRNAs (miRNAs) could play important roles in cancers via post-transcriptionally regulating target genes via binding to specific sequences in the 3' UTR of downstream target genes. In the present study, we performed microarray profiling analyses to identify differentially-expressed miRNAs in preosteoclast before and after osteoclast differentiation that could target OSM. miR-181b-5p was downregulated during Raw264.7 cells differentiation into osteoclast. By direct targeting OSM 3' UTR, miR-181b-5p inhibited OSM messenger RNA expression and protein levels, subsequently decreasing IL-6 and AREG and increasing OPG, while OSM overexpression exerted an opposing effect. More importantly, co-culture with miR-181b-5p-overexpressing differentiated Raw264.7 cells suppressed proliferation, migration, and invasion of mouse prostate cancer RM-1 cells, while co-culture with OSM-overexpressing Raw264.7 cells led to opposing cellular effects. More importantly, the effects of miR-181b-5p on osteoclastogenic factors and RM-1 cells could be significantly reversed by OSM overexpression. In summary, miR-181b-5p/OSM axis could be a viable therapeutic target for patients with surgically removed primary tumors to reduce bone metastasis and prevent bone loss.
Collapse
Affiliation(s)
- Ziwei Han
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ruisen Zhan
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shijie Chen
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia Deng
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Shi
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weiguo Wang
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
42
|
Ma W, Peltzer K, Qi L, Xu G, Liu Z, Wang J, Mao M, Chekhonin VP, Wang X, Zhang C. Female sex is associated with a lower risk of bone metastases and favourable prognosis in non-sex-specific cancers. BMC Cancer 2019; 19:1001. [PMID: 31653236 PMCID: PMC6815063 DOI: 10.1186/s12885-019-6168-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 09/16/2019] [Indexed: 11/21/2022] Open
Abstract
Background The objectives were to investigate the disparity in the prevalence of bone metastases (BM) between the sexes and to assess the effect of female sex on the development and prognosis of BM. Methods Cases of invasive non-sex-specific cancers diagnosed between 2010 and 2015 in the Surveillance, Epidemiology, and End Results (SEER) program were used. The prevalence of BM was calculated by combining the prevalence of BM among different cancers. Multivariable logistic regression and proportion hazard regression were conducted to investigate the effect of female sex, and the results were pooled by meta-analysis. Results The pooled prevalence of BM among male and female patients was 2.3% (95% CI: 1.6–3.2%) and 1.8% (95% CI: 1.2–2.6%), respectively. The pooled prevalence of BM dramatically decreased for patients aged 11–40 years old, plateaued for patients aged 41–90 years old and increased for patients aged > 90 years old in both male and female patients. Meta-analysis suggested that female sex had a protective effect on the development of BM (pooled OR = 0.80; 95% CI: 0.75–0.84; p < .001) and a favourable prognosis for respiratory system cancers (pooled HR = 0.81; 95% CI: 0.71–0.92; p < .001). However, no significant associations existed for other cancers. Male non-sex-specific cancer patients and those with male-leaning genetic variations or hormonal status have a greater likelihood of developing BM than female patients. Conclusions Female sex was associated with fewer BM in various non-sex-specific cancers, and the effect was constant with changes in age. Female sex showed a protective effect exclusively on the prognosis of respiratory system cancers.
Collapse
Affiliation(s)
- Wenjuan Ma
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Karl Peltzer
- Department of Research & Innovation, University of Limpopo, Turfloop, Mankweng, South Africa
| | - Lisha Qi
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Guijun Xu
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huanhu Xi Road, Tianjin, 300060, China
| | - Zheng Liu
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huanhu Xi Road, Tianjin, 300060, China
| | - Jingyi Wang
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Min Mao
- Department of Pathology and Southwest Cancer Center, First Affiliated Hospital, Army Medical University, Chongqing, 400038, China
| | - Vladimir P Chekhonin
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Xin Wang
- Department of Epidemiology and Biostatistics, First Affiliated Hospital, Army Medical University, 30 Gaotanyan Street, Shapingba District, 400038, China.
| | - Chao Zhang
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huanhu Xi Road, Tianjin, 300060, China.
| |
Collapse
|
43
|
Zhong P, Liu L, Shen A, Chen Z, Hu X, Cai Y, Lin J, Wang B, Li J, Chen Y, Peng J. Five extracellular matrix-associated genes upregulated in oral tongue squamous cell carcinoma: An integrated bioinformatics analysis. Oncol Lett 2019; 18:5959-5967. [PMID: 31788070 PMCID: PMC6865669 DOI: 10.3892/ol.2019.10982] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 07/26/2019] [Indexed: 12/16/2022] Open
Abstract
Despite advancements in treatment regimens, the mortality rate of patients with oral tongue squamous cell carcinoma (OTSCC) is high. In addition, the signaling pathways and oncoproteins involved in OTSCC progression remain largely unknown. Therefore, the aim of the present study was to identify specific prognostic marker for patients at a high risk of developing OTSCC. The present study used four original microarray datasets to identify the key candidate genes involved in OTSCC pathogenesis. Expression profiles of 93 OTSCC tissues and 76 normal tissues from GSE9844, GSE13601, GSE31056 and GSE75538 datasets were investigated. Differentially expressed genes (DEGs) were determined, and gene ontology enrichment and gene interactions were analyzed. The four GSE datasets reported five upregulated and six downregulated DEGs. Five upregulated genes (matrix metalloproteinase 1, 3, 10 and 12 and laminin subunit gamma 2) were localized in the extracellular region of cells and were associated with extracellular matrix disassembly. Furthermore, analysis for The Cancer Genome Atlas database revealed that the aforementioned five upregulated genes were also highly expressed in OTSCC and head and neck squamous cell carcinoma tissues. These results demonstrated that the five upregulated genes may be considered as potential prognostic biomarkers of OTSCC and may serve at understanding OTSCC progression. Upregulated DEGs may therefore represent valuable therapeutic targets to prevent or control OTSCC pathogenesis.
Collapse
Affiliation(s)
- Pingping Zhong
- Department of Orthodontics, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian 350002, P.R. China.,Key Laboratory of Stomatology, Fujian Province University, Fuzhou, Fujian 350002, P.R. China.,Fujian Biological Materials Engineering and Technology Center of Stomatology, Fuzhou, Fujian 350002, P.R. China.,Institute of Stomatology, Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Liya Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Zhongxin Chen
- West China School of Stomatology Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaoyan Hu
- Key Laboratory of Stomatology, Fujian Province University, Fuzhou, Fujian 350002, P.R. China.,Fujian Biological Materials Engineering and Technology Center of Stomatology, Fuzhou, Fujian 350002, P.R. China.,Institute of Stomatology, Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Yichao Cai
- Key Laboratory of Stomatology, Fujian Province University, Fuzhou, Fujian 350002, P.R. China.,Fujian Biological Materials Engineering and Technology Center of Stomatology, Fuzhou, Fujian 350002, P.R. China.,Institute of Stomatology, Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Jie Lin
- Key Laboratory of Stomatology, Fujian Province University, Fuzhou, Fujian 350002, P.R. China.,Fujian Biological Materials Engineering and Technology Center of Stomatology, Fuzhou, Fujian 350002, P.R. China.,Institute of Stomatology, Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Bangyan Wang
- Key Laboratory of Stomatology, Fujian Province University, Fuzhou, Fujian 350002, P.R. China.,Fujian Biological Materials Engineering and Technology Center of Stomatology, Fuzhou, Fujian 350002, P.R. China.,Institute of Stomatology, Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Jiesen Li
- Key Laboratory of Stomatology, Fujian Province University, Fuzhou, Fujian 350002, P.R. China.,Fujian Biological Materials Engineering and Technology Center of Stomatology, Fuzhou, Fujian 350002, P.R. China.,Institute of Stomatology, Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Youqin Chen
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH 44106, USA
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
44
|
Dyskova T, Kriegova E, Slobodova Z, Zehnalova S, Kudelka M, Schneiderova P, Fillerova R, Gallo J. Inflammation time-axis in aseptic loosening of total knee arthroplasty: A preliminary study. PLoS One 2019; 14:e0221056. [PMID: 31469844 PMCID: PMC6716666 DOI: 10.1371/journal.pone.0221056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 07/29/2019] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Aseptic loosening (AL) is the most frequent long-term reason for revision of total knee arthroplasty (TKA) affecting about 15-20% patients within 20 years after the surgery. Although there is a solid body of evidence about the crucial role of inflammation in the AL pathogenesis, scared information on inflammation signature and its time-axis in tissues around TKA exists. DESIGN The inflammation protein signatures in pseudosynovial tissues collected at revision surgery from patients with AL (AL, n = 12) and those with no clinical/radiographic signs of AL (non-AL, n = 9) were investigated by Proximity Extension Assay (PEA)-Immunoassay and immunohistochemistry. RESULTS AL tissues had elevated levels of TNF-family members sTNFR2, TNFSF14, sFasL, sBAFF, cytokines/chemokines IL8, CCL2, IL1RA/IL36, sIL6R, and growth factors sAREG, CSF1, comparing to non-AL. High interindividual variability in protein levels was evident particularly in non-AL. Levels of sTNFR2, sBAFF, IL8, sIL6R, and MPO discriminated between AL and non-AL and were associated with the time from index surgery, suggesting the cumulative character of inflammatory osteolytic response to prosthetic byproducts. The source of elevated inflammatory molecules was macrophages and multinucleated osteoclast-like cells in AL and histiocytes and osteoclast-like cells in non-AL tissues, respectively. All proteins were present in higher levels in osteoclast-like cells than in macrophages. CONCLUSIONS Our study revealed a differential inflammation signature between AL and non-AL stages of TKA. It also highlighted the unique patient's response to TKA in non-AL stages. Further confirmation of our preliminary results on a larger cohort is needed. Analysis of the time-axis of processes ongoing around TKA implantation may help to understand the mechanisms driving periprosthetic bone resorption needed for diagnostic/preventative strategies.
Collapse
Affiliation(s)
- Tereza Dyskova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Eva Kriegova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Zuzana Slobodova
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Sarka Zehnalova
- Department of Computer Science, Faculty of Electrical Engineering and Computer Science, VSB-Technical University of Ostrava, Ostrava, Czech Republic
| | - Milos Kudelka
- Department of Computer Science, Faculty of Electrical Engineering and Computer Science, VSB-Technical University of Ostrava, Ostrava, Czech Republic
| | - Petra Schneiderova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Regina Fillerova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jiri Gallo
- Department of Orthopaedics, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| |
Collapse
|
45
|
Li D, Wu Y, Wu Y, Ni C, Jiang P, Li J, Mao L, Zheng Q, Yue J. HtrA1 upregulates the expression of ADAMTS-5 in HNPCs via the ERK/NF-κB/JNK signaling pathway. Am J Transl Res 2019; 11:5114-5121. [PMID: 31497227 PMCID: PMC6731407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 07/30/2019] [Indexed: 06/10/2023]
Abstract
Intervertebral disc degeneration (IDD) is a form of chronic inflammation and is one of the most common disorders reported to be involved in low back pain (LBP). The pathophysiology of degeneration is not completely understood, but the consensus is that the degradation of extracellular matrix (ECM) proteins in the disc is the leading factor contributing to IDD. High temperature requirement A1 (HtrA1) is serine protease that has been shown to be increased in degenerated intervertebral discs as a result of an increase in the expression of matrix metalloproteinases (MMPs), but no study has focused on the effect of HtrA1 on a disintegrin-like and metalloproteinase with thrombospondin motifs (ADAMTSs). In the present study, we successfully isolated human nucleus pulposus cells (HNPCs) from IDD patients who were our research subjects to elaborate on the potential role of HtrA1 in the pathogenesis of IDD. We confirmed that HtrA1 has the potential to induce the expression of ADAMTS-5 in a dose-dependent manner. Consistently, this was mediated by the ERK, NF-κB and JNK pathways. By using inhibitors of these pathways, the increase in ADAMTS-5 could be reduced. Our findings indicated that HtrA1 can induce the expression of ADAMTS-5 in HNPCs via the ERK/NF-κB/JNK signaling pathway, and our study also elucidated the involved induction mechanisms in HNPCs, which may provide new insights for the treatment of IDD.
Collapse
Affiliation(s)
- Dapeng Li
- Department of Orthopedics, Affiliated Hospital of Jiangsu UniversityZhenjiang 212001, Jiangsu Province, China
| | - Yumin Wu
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow UniversityChangzhou 213003, Jiangsu Province, China
| | - Yan Wu
- Department of Physiology, Medical College of Jiangsu UniversityZhenjiang 212013, Jiangsu Province, China
| | - Chenlie Ni
- Department of Orthopedics, Affiliated Hospital of Jiangsu UniversityZhenjiang 212001, Jiangsu Province, China
| | - Pan Jiang
- Department of Orthopedics, Affiliated Hospital of Jiangsu UniversityZhenjiang 212001, Jiangsu Province, China
| | - Jian Li
- Department of Orthopedics, Affiliated Hospital of Jiangsu UniversityZhenjiang 212001, Jiangsu Province, China
| | - Lianghao Mao
- Department of Orthopedics, Affiliated Hospital of Jiangsu UniversityZhenjiang 212001, Jiangsu Province, China
| | - Qiping Zheng
- Jiangsu Key Laboratory of Medical Science and The Laboratory of Jiangsu UniversityZhenjiang 212013, Jiangsu Province, China
| | - Jiawei Yue
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow UniversityChangzhou 213003, Jiangsu Province, China
| |
Collapse
|
46
|
Liang Y, Zhang H, Song X, Yang Q. Metastatic heterogeneity of breast cancer: Molecular mechanism and potential therapeutic targets. Semin Cancer Biol 2019; 60:14-27. [PMID: 31421262 DOI: 10.1016/j.semcancer.2019.08.012] [Citation(s) in RCA: 468] [Impact Index Per Article: 93.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 08/11/2019] [Accepted: 08/12/2019] [Indexed: 02/08/2023]
Abstract
Breast cancer is one of the most common malignancies among women throughout the world and is the major cause of most cancer-related deaths. Several explanations account for the high rate of mortality of breast cancer, and metastasis to vital organs is identified as the principal cause. Over the past few years, intensive efforts have demonstrated that breast cancer exhibits metastatic heterogeneity with distinct metastatic precedence to various organs, giving rise to differences in prognoses and responses to therapy in breast cancer patients. Bone, lung, liver, and brain are generally accepted as the primary target sites of breast cancer metastasis. However, the underlying molecular mechanism of metastatic heterogeneity of breast cancer remains to be further elucidated. Recently, the advent of novel genomic and pathologic approaches as well as technological breakthroughs in imaging analysis and animal modelling have yielded an unprecedented change in our understanding of the heterogeneity of breast cancer metastasis and provided novel insight for establishing more effective therapeutics. This review summarizes recent molecular mechanisms and emerging concepts on the metastatic heterogeneity of breast cancer and discusses the potential of identifying specific molecules against tumor cells or tumor microenvironments to thwart the development of metastatic disease and improve the prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Yiran Liang
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, PR China
| | - Hanwen Zhang
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaojin Song
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, PR China
| | - Qifeng Yang
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, PR China; Pathology Tissue Bank, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
47
|
Wei YF, Huang WT, Liu TC, Shieh JM, Chian CF, Wu MF, Chang CC, Lin CH, Ko JC, Lin CM, Hsia TC. Factors associated with improvement in symptoms and quality of life for first-line EGFR-tyrosine kinase inhibitor treatment in patients with EGFR-mutated non-small-cell lung cancer - A multicenter prospective SMILE study. J Cancer 2019; 10:4151-4158. [PMID: 31417660 PMCID: PMC6692623 DOI: 10.7150/jca.30507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 04/28/2019] [Indexed: 12/17/2022] Open
Abstract
Introduction: Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) are a standard first-line treatment for advanced EGFR-mutated NSCLC patients. Factors associated with symptoms and quality of life (QOL) improvements have not been investigated. Methods: We conducted a multicenter, prospective study to evaluate improvements in QOL and symptoms in NSCLC patients treated with first-line EGFR-TKIs. QOL was assessed using the instrument of Functional Assessment of Cancer Therapy-Lung questionnaire (FACT-L) and Treatment Outcome Index (TOI). Assessment of symptoms was evaluated using the Lung cancer subscale (LCS). Results: Eligible subjects included 280 patients for endpoint analyses. The mean FACT-L score increased by 4.0 ± 15.56 at Week 2 (p<0.001), 5.1 ± 18.48 at Week 4 (p<0.001), and 4.2 ± 20.27 at Week 12 (p=0.001). Similarly, a 2.3 ± 11.65 (p<0.001), 3.2 ± 13.59 (p<0.001), and 2.4 ± 14.34 (p=0.009) increase in mean TOI score were observed at Weeks 2, 4 and 12, respectively. For LCS, it was slightly increased by 1.7 ± 4.61, 2.0 ± 5.50, and 2.0 ± 5.36 at Weeks 2, 4, and 12 (all p<0.001), respectively. Subgroup analyses showed patients who were ex-smokers or with at least 3 metastatic sites were associated with symptoms improvement. Patients who were ex-smokers, with at least 3 metastatic sites, a PS of 1, or treated with gefitinib were associated with QOL improvement. Conclusions: In EGFR -mutated NSCLC patients who were treated with first-line EGFR-TKIs, these ex-smokers or with 3 or more metastatic sites were associated with improvements in symptoms and QOL.
Collapse
Affiliation(s)
- Yu-Feng Wei
- Division of Chest Department, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.,Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung, Taiwan
| | - Wen-Tsung Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Chi Mei Medical Center Liouying, Tainan, Taiwan
| | - Tu-Chen Liu
- Department of Chest Medicine, Cheng Ching Hospital-Chung Kang Branch, Taichung, Taiwan
| | - Jiunn-Min Shieh
- Department of Internal Medicine, Chi Mei Medical Center - YongKang Branch, Tainan, Taiwan
| | - Chih-Feng Chian
- Division of Pulmonary and Critical Care Medicine, Internal Medicine Department, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Fang Wu
- Division of Medical Oncology, Department of Internal Medicine, Chung Shan Medical University Hospital, and School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chih-Cheng Chang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University
| | - Ching-Hsiung Lin
- Division of Chest Medicine, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Jen-Chung Ko
- Department of Internal Medicine, National Taiwan University Hospital - Hsinchu Branch, Hsinchu, Taiwan
| | - Chia-Mo Lin
- Sleep Center, Pulmonary and Critical Care Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Te-Chun Hsia
- Department of Respiratory Therapy, China Medical University, Taichung, Taiwan.,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
48
|
Pan-cancer analysis connects tumor matrisome to immune response. NPJ Precis Oncol 2019; 3:15. [PMID: 31123708 PMCID: PMC6531473 DOI: 10.1038/s41698-019-0087-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 04/10/2019] [Indexed: 01/10/2023] Open
Abstract
Recent sequencing efforts unveil genomic landscapes of tumor microenvironment. A key compartment in this niche is the extracellular matrix (ECM) and its related components - matrisome. Yet, little is known about the extent to which matrisome pattern is conserved in progressive tumors across diverse cancer types. Using integrative genomic approaches, we conducted multi-platform assessment of a measure of deregulated matrisome associated with tumor progression, termed as tumor matrisome index (TMI), in over 30,000 patient-derived samples. Combined quantitative analyses of genomics and proteomics reveal that TMI is closely associated with mutational load, tumor pathology, and predicts survival across different malignancies. Interestingly, we observed an enrichment of specific tumor-infiltrating immune cell populations, along with signatures predictive of resistance to immune checkpoint blockade immunotherapy, and clinically targetable immune checkpoints in TMIhigh tumors. B7-H3 emerged as a particularly promising target for anti-tumor immunity in these tumors. Here, we show that matrisomal abnormalities could represent a potential clinically useful biomarker for prognostication and prediction of immunotherapy response.
Collapse
|
49
|
Liu R, Zhong J, Yu X, Li Y, Chen P. Identifying Critical State of Complex Diseases by Single-Sample-Based Hidden Markov Model. Front Genet 2019; 10:285. [PMID: 31019526 PMCID: PMC6458292 DOI: 10.3389/fgene.2019.00285] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/15/2019] [Indexed: 12/20/2022] Open
Abstract
The progression of complex diseases is generally divided as a normal state, a pre-disease state or tipping point, and a disease state. Developing individual-specific method that can identify the pre-disease state just before a catastrophic deterioration, is critical for patients with complex diseases. However, with only a case sample, it is challenging to detect a pre-disease state which has little significant differences comparing with a normal state in terms of phenotypes and gene expressions. In this study, by regarding the tipping point as the end point of a stationary Markov process, we proposed a single-sample-based hidden Markov model (HMM) approach to explore the dynamical differences between a normal and a pre-disease states, and thus can signal the upcoming critical transition immediately after a pre-disease state. Using this method, we identified the pre-disease state or tipping point in a numerical simulation and two real datasets including stomach adenocarcinoma and influenza infection, which demonstrate the effectiveness of the method.
Collapse
Affiliation(s)
- Rui Liu
- School of Mathematics, South China University of Technology, Guangzhou, China
| | - Jiayuan Zhong
- School of Mathematics, South China University of Technology, Guangzhou, China
| | - Xiangtian Yu
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yongjun Li
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, China
| | - Pei Chen
- School of Mathematics, South China University of Technology, Guangzhou, China
| |
Collapse
|
50
|
Li P, Deng Q, Liu J, Yan J, Wei Z, Zhang Z, Liu H, Li B. Roles for HB-EGF in Mesenchymal Stromal Cell Proliferation and Differentiation During Skeletal Growth. J Bone Miner Res 2019; 34:295-309. [PMID: 30550637 PMCID: PMC7816091 DOI: 10.1002/jbmr.3596] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 09/04/2018] [Accepted: 09/22/2018] [Indexed: 12/22/2022]
Abstract
HB-EGF, a member of the EGF superfamily, plays important roles in development and tissue regeneration. However, its functions in skeletal stem cells and skeleton development and growth remain poorly understood. Here, we used the Cre/LoxP system to ablate or express HB-EGF in Dermo1+ mesenchymal stromal cells and their progenies, including chondrocytes and osteoblast lineage cells, and bone marrow stromal cells (BMSCs). Dermo1-Cre; HB-EGFf/f mice only showed a modest increase in bone mass, whereas Dermo1-HB-EGF mice developed progressive chondrodysplasia, chondroma, osteoarthritis-like joint defects, and loss of bone mass and density, which were alleviated by treatment with EGFR inhibitor AG1478. The cartilage defects were recapitulated in chondrocyte-specific HB-EGF overexpression (Col2-HB-EGF) mice with a lesser severity. Dermo1-HB-EGF mice showed an increase in proliferation but defects in differentiation of chondrocytes and osteoblasts. HB-EGF promoted BMSC proliferation via the Akt1 and Erk pathways but inhibited BMSC differentiation via restraining Smad1/5/8 activation. However, Dermo1-HB-EGF mice showed normal osteoclastogenesis and bone resorption. These results reveal an important function of autocrine or paracrine HB-EGF in mesenchymal stromal cell proliferation and differentiation and suggest that EGF signaling needs to be tightly controlled to maintain bone and articular cartilage integrity. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
- Ping Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Deng
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Jiajia Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Jianshe Yan
- School of Life Sciences, Shanghai University, Shanghai, China.,Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhanying Wei
- Metabolic Bone Disease and Genetic Research Unit, Department of Osteoporosis and Bone Diseases, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhenlin Zhang
- Metabolic Bone Disease and Genetic Research Unit, Department of Osteoporosis and Bone Diseases, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|