1
|
Khaleel AQ, Altalbawy FMA, Jabir MS, F Hasan T, Jain V, Abbot V, Nakash P, Kumar MR, Mustafa YF, Jawad MA. CXCR4/CXCL12 blockade therapy; a new horizon in TNBC therapy. Med Oncol 2025; 42:161. [PMID: 40216617 DOI: 10.1007/s12032-025-02705-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/29/2025] [Indexed: 05/03/2025]
Abstract
The only subtype of breast cancer (BC) without specific therapy is triple-negative breast cancer (TNBC), which represents 15-20% of incidence cases of BC. TNBC encompasses transformed and nonmalignant cells, including cancer-associated fibroblasts (CAF), endothelial vasculature, and tumor-infiltrating cells. These nonmalignant cells, soluble factors (e.g., cytokines), and the extracellular matrix (ECM) form the tumor microenvironment (TME). The TME is made up of these nonmalignant cells, ECM, and soluble components, including cytokines. Direct cell-to-cell contact and soluble substances like cytokines (e.g., chemokines) may facilitate interaction between cancer cells and the surrounding TME. Through growth-promoting cytokines, TME not only enables the development of cancer but also confers therapy resistance. New treatment targets will probably be suggested by comprehending the processes behind tumor development and progression as well as the functions of chemokines in TNBC. In this light, several investigations have shown the pivotal function of the C-X-C motif chemokine ligand 12 (CXCL12 or SDF-1) axis and chemokine receptor type 4 (CXCR4) in the pathophysiology of TNBC. This review provides an overview of the CXCR4/CXCL12 axis' function in TNBC development, metastasis, angiogenesis, and treatment resistance. A synopsis of current literature on targeting the CXCR4/CXCL12 axis for treating and managing TNBC has also been provided.
Collapse
Affiliation(s)
- Abdulrahman Qais Khaleel
- Department of Medical Instruments Engineering, Al-Maarif University College, Al Anbar, 31001, Iraq.
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Majid S Jabir
- Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Thikra F Hasan
- College of Health&Medical Technology, Uruk University, Baghdad, Iraq
| | - Vicky Jain
- Department of Chemistry, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Vikrant Abbot
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Prashant Nakash
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | | |
Collapse
|
2
|
Horiguchi H, Kadomatsu T, Oike Y. The Two Faces of Angiopoietin-Like Protein 2 in Cancer. Cancer Sci 2025; 116:592-599. [PMID: 39686837 PMCID: PMC11875762 DOI: 10.1111/cas.16434] [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: 11/01/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 12/18/2024] Open
Abstract
The tumor microenvironment is composed of tumor cells and various stromal cell types, such as immune cells, fibroblasts, and vascular cells. Signaling interactions between tumor and stromal cells orchestrate the tumor microenvironment's contribution to tumor progression. Angiopoietin-like protein 2 (ANGPTL2) is a secreted glycoprotein homologous to angiopoietins. Previous studies indicate that tumor cell-derived ANGPTL2 serves as a tumor promoter. However, recent studies suggest that tumor stroma-derived ANGPTL2 shows tumor-suppressive activity by enhancing anti-tumor immune responses, supporting a dual function for ANGPTL2 in cancer pathology. Such complexity can complicate development of effective therapeutic strategies targeting ANGPTL2. In this Review, we focus on ANGPTL2 activity in the tumor microenvironment and its function in anti-cancer immunity.
Collapse
Affiliation(s)
- Haruki Horiguchi
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Aging and Geriatric Medicine, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Aging and Geriatric Medicine, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
3
|
Thilakan J, Goel SK, Arya N. In-situ collagen mineralization modulates metastatic properties of breast cancer cells. J Biosci Bioeng 2025; 139:123-132. [PMID: 39580239 DOI: 10.1016/j.jbiosc.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 06/20/2024] [Accepted: 07/12/2024] [Indexed: 11/25/2024]
Abstract
Bone metastasis is the leading cause of morbidity and mortality in advanced-stage breast cancer patients. While most studies focus on the cellular and genetic factors associated with breast cancer metastasis, the role of the extracellular matrix (ECM) of bone in breast cancer metastasis remains elusive. In this study, we recapitulated the bone microenvironment using in-situ mineralized collagen type-I hydrogels and utilized them to understand breast cancer metastasis. Our results indicated successful mineralization of collagen type-I based hydrogels in the presence of serum proteins, which increased as a function of time. There was no difference in the adhesion of breast cancer cells seeded on collagen and mineralized collagen surfaces. However, there was a marked reduction in cell proliferation, down-regulation of various metastatic markers, and decreased migratory phenotype with a concomitant increase in cleaved caspase-3 on mineralized collagen compared to collagen hydrogels. In conclusion, our results suggest an inverse relationship between bone mineralization and the metastatic propensity of breast cancer cells. We further speculate the role of other factors in the skeletal ecosystem for mediating preferential homing of breast cancer cells to the bone microenvironment.
Collapse
Affiliation(s)
- Jaya Thilakan
- Department of Biochemistry, All India Institute of Medical Sciences Bhopal, Bhopal 462020, Madhya Pradesh, India; Department of Genetics, UTD, Barkatullah University Bhopal, Bhopal 462026, Madhya Pradesh, India
| | - Sudhir Kumar Goel
- Department of Biochemistry, All India Institute of Medical Sciences Bhopal, Bhopal 462020, Madhya Pradesh, India; Department of Biochemistry, T. S. Misra Medical College and Hospital, Amousi, Lucknow 226008, Uttar Pradesh, India
| | - Neha Arya
- Department of Translational Medicine, All India Institute of Medical Sciences Bhopal, Bhopal 462020, Madhya Pradesh, India.
| |
Collapse
|
4
|
Inson I, Chutoe C, Kanjanapipak J, Lertsuwan K. Cannabinoid Receptor Type 2 Agonist, GW405833, Reduced the Impacts of MDA-MB-231 Breast Cancer Cells on Bone Cells. Cancer Med 2025; 14:e70709. [PMID: 39980332 PMCID: PMC11842928 DOI: 10.1002/cam4.70709] [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: 11/16/2024] [Revised: 01/22/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
AIM Breast cancer frequently metastasizes to bones. The interaction between breast cancer cells and bone cells results in osteolytic lesions by disrupting the balance between osteoblast-mediated bone production and osteoclast-mediated bone resorption. This study aims to investigate the effects of the cannabinoid receptor type 2 (CB2) agonist, GW405833, on interactions between breast cancer cells and osteoblasts as well as its impact on breast cancer-induced osteoclastogenesis. MATERIALS & METHODS MDA-MB-231, UMR-106, RAW 264.7 cells were used to represent breast cancer cells, osteoblast-like cells and macrophage-osteoclast precursor cells, respectively. Cell viability was evaluated by MTT assay, and breast cancer cell invasion was assessed by Transwell invasion assay. Tartrate-resistant acid phosphatase (TRAP) staining was utilized to evaluate osteoclastogenesis. RESULTS Our results demonstrated that GW405833 disrupted MDA-MB-231-induced UMR-106 cell death and promoted UMR-106 cell viability. The underlying mechanism of these effects was determined in this study. GW405833 reduced AKT phosphorylation in MDA-MB-231 cells without affecting mTOR protein expression or its phosphorylation. Conversely, in UMR-106 cells, GW405833 induced AKT and mTOR phosphorylated protein. Furthermore, the mTOR inhibitor reversed the GW405833-induced recovery of UMR-106 cell viability under MDA-MB-231-derived conditioned media (CM) exposure. These findings underscore the critical role of the AKT/mTOR pathway in mediating GW405833's inhibitory effects on cancer-bone interactions. Additionally, GW405833 suppressed osteoblast-enhanced breast cancer cell invasion and the expression of invasion-related proteins in both cell types, along with reducing osteoclastogenic factors induced by MDA-MB-231 CM in UMR-106 cells and suppressing MDA-MB-231 CM-enhanced osteoclastogenesis in RAW 264.7 cells. CONCLUSION This study highlights the therapeutic potential of cannabinoid receptor agonist for treating breast cancer bone metastasis and bone-related complications.
Collapse
Affiliation(s)
- Ingon Inson
- Department of Biochemistry, Faculty of ScienceMahidol UniversityBangkokThailand
| | - Chartinun Chutoe
- Department of Biochemistry, Faculty of ScienceMahidol UniversityBangkokThailand
| | | | - Kornkamon Lertsuwan
- Department of Biochemistry, Faculty of ScienceMahidol UniversityBangkokThailand
- Center of Calcium and Bone Research (COCAB), Faculty of ScienceMahidol UniversityBangkokThailand
| |
Collapse
|
5
|
Wang Y, Hu Y, Wang M, Wang M, Xu Y. The Role of Breast Cancer Cells in Bone Metastasis: Suitable Seeds for Nourishing Soil. Curr Osteoporos Rep 2024; 22:28-43. [PMID: 38206556 DOI: 10.1007/s11914-023-00849-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 01/12/2024]
Abstract
PURPOSE OF REVIEW The purpose of this review was to describe the characteristics of breast cancer cells prone to developing bone metastasis and determine how they are regulated by the bone microenvironment. RECENT FINDINGS The bone is a site of frequent breast cancer metastasis. Bone metastasis accounts for 70% of advanced breast cancer cases and remains incurable. It can lead to skeletal-related events, such as bone fracture and pain, and seriously affect the quality of life of patients. Breast cancer cells escape from the primary lesion and spread to the bone marrow in the early stages. They can then enter the dormant state and restore tumourigenicity after several years to develop overt metastasis. In the last few years, an increasing number of studies have reported on the factors promoting bone metastasis of breast cancer cells, both at the primary and metastatic sites. Identifying factors associated with bone metastasis aids in the early recognition of bone metastasis tendency. How to target these factors and minimize the side effects on the bone remains to be further explored.
Collapse
Affiliation(s)
- Yiou Wang
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Hu
- Department of Outpatient, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mozhi Wang
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mengshen Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yingying Xu
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
6
|
Ibragimova MK, Tsyganov MM, Kravtsova EA, Tsydenova IA, Litviakov NV. Organ-Specificity of Breast Cancer Metastasis. Int J Mol Sci 2023; 24:15625. [PMID: 37958607 PMCID: PMC10650169 DOI: 10.3390/ijms242115625] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Breast cancer (BC) remains one of the most common malignancies among women worldwide. Breast cancer shows metastatic heterogeneity with priority to different organs, which leads to differences in prognosis and response to therapy among patients. The main targets for metastasis in BC are the bone, lung, liver and brain. The molecular mechanism of BC organ-specificity is still under investigation. In recent years, the appearance of new genomic approaches has led to unprecedented changes in the understanding of breast cancer metastasis organ-specificity and has provided a new platform for the development of more effective therapeutic agents. This review summarises recent data on molecular organ-specific markers of metastasis as the basis of a possible therapeutic approach in order to improve the diagnosis and prognosis of patients with metastatically heterogeneous breast cancer.
Collapse
Affiliation(s)
- Marina K. Ibragimova
- Department of Experimental Oncology, Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634009, Russia; (M.M.T.); (E.A.K.); (I.A.T.); (N.V.L.)
- Biological Institute, National Research Tomsk State University, Tomsk 634050, Russia
- Faculty of Medicine and Biology, Siberian State Medical University, Tomsk 634050, Russia
| | - Matvey M. Tsyganov
- Department of Experimental Oncology, Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634009, Russia; (M.M.T.); (E.A.K.); (I.A.T.); (N.V.L.)
- Faculty of Medicine and Biology, Siberian State Medical University, Tomsk 634050, Russia
| | - Ekaterina A. Kravtsova
- Department of Experimental Oncology, Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634009, Russia; (M.M.T.); (E.A.K.); (I.A.T.); (N.V.L.)
- Biological Institute, National Research Tomsk State University, Tomsk 634050, Russia
| | - Irina A. Tsydenova
- Department of Experimental Oncology, Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634009, Russia; (M.M.T.); (E.A.K.); (I.A.T.); (N.V.L.)
- Biological Institute, National Research Tomsk State University, Tomsk 634050, Russia
| | - Nikolai V. Litviakov
- Department of Experimental Oncology, Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634009, Russia; (M.M.T.); (E.A.K.); (I.A.T.); (N.V.L.)
- Biological Institute, National Research Tomsk State University, Tomsk 634050, Russia
- Faculty of Medicine and Biology, Siberian State Medical University, Tomsk 634050, Russia
| |
Collapse
|
7
|
Liu X, Qin J, Nie J, Gao R, Hu S, Sun H, Wang S, Pan Y. ANGPTL2+cancer-associated fibroblasts and SPP1+macrophages are metastasis accelerators of colorectal cancer. Front Immunol 2023; 14:1185208. [PMID: 37691929 PMCID: PMC10483401 DOI: 10.3389/fimmu.2023.1185208] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
Background Liver metastasis (LM) is a leading cause of cancer-related deaths in CRC patients, whereas the associated mechanisms have not yet been fully elucidated. Therefore, it is urgently needed to deeply explore novel metastasis accelerators and therapeutic targets of LM-CRC. Methods The bulk RNA sequencing data and clinicopathological information of CRC patients were enrolled from the TCGA and GEO databases. The single-cell RNA sequencing (scRNA-seq) datasets of CRC were collected from and analyzed in the Tumor Immune Single-cell Hub (TISCH) database. The infiltration levels of cancer-associated fibroblasts (CAFs) and macrophages in CRC tissues were estimated by multiple immune deconvolution algorithms. The prognostic values of genes were identified by the Kaplan-Meier curve with a log-rank test. GSEA analysis was carried out to annotate the significantly enriched gene sets. The biological functions of cells were experimentally verified. Results In the present study, hundreds of differentially expressed genes (DEGs) were selected in LM-CRC compared to primary CRC, and these DEGs were significantly associated with the regulation of endopeptidase activity, blood coagulation, and metabolic processes. Then, SPP1, CAV1, ANGPTL2, and COLEC11 were identified as the characteristic DEGs of LM-CRC, and higher expression levels of SPP1 and ANGPTL2 were significantly associated with worse clinical outcomes of CRC patients. In addition, ANGPTL2 and SPP1 mainly distributed in the tumor microenvironment (TME) of CRC tissues. Subsequent scRNA-seq analysis demonstrated that ANGPTL2 and SPP1 were markedly enriched in the CAFs and macrophages of CRC tissues, respectively. Moreover, we identified the significantly enriched gene sets in LM-CRC, especially those in the SPP1+macrophages and ANGPTL2+CAFs, such as the HALLMARK_EPITHELIAL_MESENCHYMAL_TRANSITION and the HALLMARK_COMPLEMENT. Finally, our in vitro experiments proved that ANGPTL2+CAFs and SPP1+macrophages promote the metastasis of CRC cells. Conclusion Our study selected four characteristic genes of LM-CRC and identified ANGPTL2+CAFs and SPP1+macrophages subtypes as metastasis accelerators of CRC which provided a potential therapeutic target for LM-CRC.
Collapse
Affiliation(s)
- Xiangxiang Liu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jian Qin
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junjie Nie
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Gao
- Division of Clinical Pharmacy, General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Shangshang Hu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Huiling Sun
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shukui Wang
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuqin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
8
|
Motyka J, Gacuta E, Kicman A, Kulesza M, Malinowski P, Ławicki S. CXCL12 and CXCR4 as Potential Early Biomarkers for Luminal A and Luminal B Subtypes of Breast Cancer. Cancer Manag Res 2023; 15:573-589. [PMID: 37426394 PMCID: PMC10329441 DOI: 10.2147/cmar.s416382] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 06/15/2023] [Indexed: 07/11/2023] Open
Abstract
Purpose Breast cancer is the most common type of malignancy in women. Factors that increase the risk of occurrence include chronic inflammation, with chemokines as its mediators. Therefore, the purpose of the present study was to determine the diagnostic utility of CXCL12 and CXCR4 as modern tumor markers in patients with early-stage luminal A and luminal B subtype of breast cancer and also to compare the results with the routinely used marker - CA 15-3. Patients and Methods The study included 100 patients with early breast cancer of luminal A and B subtypes, 50 women with benign breast lesion and 50 healthy women. The levels of CXCL12 and CXCR4 concentrations were determined by enzyme-linked immunosorbent assay (ELISA), comparative marker CA 15-3 - by electrochemiluminescence method (ECLIA). Results Concentrations of CXCL12 were significantly lower, while CXCR4 and CA 15-3 - significantly higher among patients with early-stage breast cancer than healthy women. CXCL12 also showed lower concentrations among fibroadenoma patients in comparison to healthy women, while CXCR4 - lower concentrations among fibroadenoma patients than cancer group. CXCL12 showed significantly higher values of sensitivity (79%), specificity (82%), positive predictive value (89.72%), negative predictive value (80%), diagnostic accuracy (80%) and diagnostic power (AUC = 0.8196) in the whole breast cancer group compared to the CA 15-3 marker (58%; 72%; 80.56%; 46.15%, 62.67%, 0.6434, resp.). Analysis of combined parameters resulted in increased sensitivity, negative predictive value and power of the test with a slight decrease in positive predictive value and a more significant decrease in specificity, reaching the best values for the three-parameter test CXCL12+CXCR4+CA15-3 (96%; 85.71%; AUC = 0.8812; 78.69%; 48%, resp.). Conclusion The results indicate the preliminary usefulness of CXCL12 and CXCR4 as early biomarkers in the diagnosis of breast cancer, especially in the combined panel with CA 15-3.
Collapse
Affiliation(s)
- Joanna Motyka
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Ewa Gacuta
- Department of Perinatology, University Clinical Hospital of Bialystok, Bialystok, Poland
| | - Aleksandra Kicman
- Department of Aesthetic Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Monika Kulesza
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Paweł Malinowski
- Department of Oncological Surgery, Bialystok Oncology Center, Bialystok, Poland
| | - Sławomir Ławicki
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
9
|
Johnson CS, Cook LM. Osteoid cell-derived chemokines drive bone-metastatic prostate cancer. Front Oncol 2023; 13:1100585. [PMID: 37025604 PMCID: PMC10070788 DOI: 10.3389/fonc.2023.1100585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
One of the greatest challenges in improving prostate cancer (PCa) survival is in designing new therapies to effectively target bone metastases. PCa regulation of the bone environment has been well characterized; however, bone-targeted therapies have little impact on patient survival, demonstrating a need for understanding the complexities of the tumor-bone environment. Many factors contribute to creating a favorable microenvironment for prostate tumors in bone, including cell signaling proteins produced by osteoid cells. Specifically, there has been extensive evidence from both past and recent studies that emphasize the importance of chemokine signaling in promoting PCa progression in the bone environment. Chemokine-focused strategies present promising therapeutic options for treating bone metastasis. These signaling pathways are complex, with many being produced by (and exerting effects on) a plethora of different cell types, including stromal and tumor cells of the prostate tumor-bone microenvironment. This review highlights an underappreciated molecular family that should be interrogated for treatment of bone metastatic prostate cancer (BM-PCa).
Collapse
Affiliation(s)
- Catherine S. Johnson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, NE, United States
| | - Leah M. Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Leah M. Cook,
| |
Collapse
|
10
|
Wu Y, Li X, Li Q, Cheng C, Zheng L. Adipose tissue-to-breast cancer crosstalk: Comprehensive insights. Biochim Biophys Acta Rev Cancer 2022; 1877:188800. [PMID: 36103907 DOI: 10.1016/j.bbcan.2022.188800] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
The review focuses on mechanistic evidence for the link between obesity and breast cancer. According to the IARC study, there is sufficient evidence that obesity is closely related to a variety of cancers. Among them, breast cancer is particularly disturbed by adipose tissue due to the unique histological structure of the breast. The review introduces the relationship between obesity and breast cancer from two aspects, including factors that promote tumorigenesis or metastasis. We summarize alterations in adipokines and metabolic pathways that contribute to breast cancer development. Breast cancer metastasis is closely related to obesity-induced pro-inflammatory microenvironment, adipose stem cells, and miRNAs. Based on the mechanism by which obesity causes breast cancer, we list possible therapeutic directions, including reducing the risk of breast cancer and inhibiting the progression of breast cancer. We also discussed the risk of autologous breast remodeling and fat transplantation. Finally, the causes of the obesity paradox and the function of enhancing immunity are discussed. Evaluating the balance between obesity-induced inflammation and enhanced immunity warrants further study.
Collapse
Affiliation(s)
- Yuan Wu
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Xu Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Qiong Li
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Chienshan Cheng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Lan Zheng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China.
| |
Collapse
|
11
|
Horiguchi H, Kadomatsu T, Yumoto S, Masuda T, Miyata K, Yamamura S, Sato M, Morinaga J, Ohtsuki S, Baba H, Moroishi T, Oike Y. Tumor cell-derived ANGPTL2 promotes β-catenin-driven intestinal tumorigenesis. Oncogene 2022; 41:4028-4041. [PMID: 35831580 DOI: 10.1038/s41388-022-02405-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 11/09/2022]
Abstract
Uncontrolled proliferation of intestinal epithelial cells caused by mutations in genes of the WNT/β-catenin pathway is associated with development of intestinal cancers. We previously reported that intestinal stromal cell-derived angiopoietin-like protein 2 (ANGPTL2) controls epithelial regeneration and intestinal immune responses. However, the role of tumor cell-derived ANGPTL2 in intestinal tumorigenesis remained unclear. Here, we show that tumor cell-derived ANGPTL2 promotes β-catenin-driven intestinal tumorigenesis. ANGPTL2 deficiency suppressed intestinal tumor development in an experimental mouse model of sporadic colon cancer. We also found that increased ANGPTL2 expression in colorectal cancer (CRC) cells augments β-catenin pathway signaling and promotes tumor cell proliferation. Relevant to mechanism, our findings suggest that tumor cell-derived ANGPTL2 upregulates expression of OB-cadherin, which then interacts with β-catenin, blocking destruction complex-independent proteasomal degradation of β-catenin proteins. Moreover, our observations support a model whereby ANGPTL2-induced OB-cadherin expression in CRC cells is accompanied by decreased cell surface integrin α5β1 expression. These findings overall provide novel insight into mechanisms of β-catenin-driven intestinal tumorigenesis.
Collapse
Affiliation(s)
- Haruki Horiguchi
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan.,Department of Aging and Geriatric Medicine, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan. .,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
| | - Shinsei Yumoto
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan.,Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Takeshi Masuda
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Keishi Miyata
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Shuji Yamamura
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Michio Sato
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Jun Morinaga
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Toshiro Moroishi
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.,Department of Cell Signaling and Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan. .,Department of Aging and Geriatric Medicine, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan. .,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
| |
Collapse
|
12
|
Kim BK, Kim DM, Park H, Kim SK, Hwang MA, Lee J, Kang MJ, Byun JE, Im JY, Kang M, Park KC, Yeom YI, Kim SY, Jung H, Kweon DH, Cheong JH, Won M. Synaptotagmin 11 scaffolds MKK7-JNK signaling process to promote stem-like molecular subtype gastric cancer oncogenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:212. [PMID: 35768842 PMCID: PMC9241269 DOI: 10.1186/s13046-022-02420-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/14/2022] [Indexed: 12/15/2022]
Abstract
Background Identifying biomarkers related to the diagnosis and treatment of gastric cancer (GC) has not made significant progress due to the heterogeneity of tumors. Genes involved in histological classification and genetic correlation studies are essential to develop an appropriate treatment for GC. Methods In vitro and in vivo lentiviral shRNA library screening was performed. The expression of Synaptotagmin (SYT11) in the tumor tissues of patients with GC was confirmed by performing Immunohistochemistry, and the correlation between the expression level and the patient’s survival rate was analyzed. Phospho-kinase array was performed to detect Jun N-terminal kinase (JNK) phosphorylation. SYT11, JNK, and MKK7 complex formation was confirmed by western blot and immunoprecipitation assays. We studied the effects of SYT11 on GC proliferation and metastasis, real-time cell image analysis, adhesion assay, invasion assay, spheroid formation, mouse xenograft assay, and liver metastasis. Results SYT11 is highly expressed in the stem-like molecular subtype of GC in transcriptome analysis of 527 patients with GC. Moreover, SYT11 is a potential prognostic biomarker for histologically classified diffuse-type GC. SYT11 functions as a scaffold protein, binding both MKK7 and JNK1 signaling molecules that play a role in JNK1 phosphorylation. In turn, JNK activation leads to a signaling cascade resulting in cJun activation and expression of downstream genes angiopoietin-like 2 (ANGPTL2), thrombospondin 4 (THBS4), Vimentin, and junctional adhesion molecule 3 (JAM3), which play a role in epithelial-mesenchymal transition (EMT). SNU484 cells infected with SYT11 shRNA (shSYT11) exhibited reduced spheroid formation, mouse tumor formation, and liver metastasis, suggesting a pro-oncogenic role of SYT11. Furthermore, SYT11-antisense oligonucleotide (ASO) displayed antitumor activity in our mouse xenograft model and was conferred an anti-proliferative effect in SNU484 and MKN1 cells. Conclusion SYT11 could be a potential therapeutic target as well as a prognostic biomarker in patients with diffuse-type GC, and SYT11-ASO could be used in therapeutic agent development for stem-like molecular subtype diffuse GC. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02420-3.
Collapse
Affiliation(s)
- Bo-Kyung Kim
- Personalized Genomic Medicine Research Center, KRIBB, 125 Kwahag-ro, Yuseong-gu, Daejeon, 34141, South Korea. .,KRIBB School of Bioscience, University of Science and Technology, Daejeon, South Korea. .,R&D Center, oneCureGEN, Daejeon, South Korea.
| | - Da-Mi Kim
- Personalized Genomic Medicine Research Center, KRIBB, 125 Kwahag-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Hyunkyung Park
- Personalized Genomic Medicine Research Center, KRIBB, 125 Kwahag-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Seon-Kyu Kim
- Personalized Genomic Medicine Research Center, KRIBB, 125 Kwahag-ro, Yuseong-gu, Daejeon, 34141, South Korea.,KRIBB School of Bioscience, University of Science and Technology, Daejeon, South Korea
| | - Mi-Aie Hwang
- Personalized Genomic Medicine Research Center, KRIBB, 125 Kwahag-ro, Yuseong-gu, Daejeon, 34141, South Korea.,Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Jungwoon Lee
- KRIBB School of Bioscience, University of Science and Technology, Daejeon, South Korea.,Environmental Diseases Research Center, KRIBB, Daejeon, South Korea
| | - Mi-Jung Kang
- Personalized Genomic Medicine Research Center, KRIBB, 125 Kwahag-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Jae-Eun Byun
- Immunotherapy Research Center, KRIBB, Daejeon, South Korea
| | - Joo-Young Im
- Personalized Genomic Medicine Research Center, KRIBB, 125 Kwahag-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Minho Kang
- Personalized Genomic Medicine Research Center, KRIBB, 125 Kwahag-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Kyung Chan Park
- Personalized Genomic Medicine Research Center, KRIBB, 125 Kwahag-ro, Yuseong-gu, Daejeon, 34141, South Korea.,KRIBB School of Bioscience, University of Science and Technology, Daejeon, South Korea
| | - Young Il Yeom
- Personalized Genomic Medicine Research Center, KRIBB, 125 Kwahag-ro, Yuseong-gu, Daejeon, 34141, South Korea.,KRIBB School of Bioscience, University of Science and Technology, Daejeon, South Korea
| | - Seon-Young Kim
- KRIBB School of Bioscience, University of Science and Technology, Daejeon, South Korea.,Korea Bioinformation Center, KRIBB, Daejeon, South Korea
| | - Haiyoung Jung
- KRIBB School of Bioscience, University of Science and Technology, Daejeon, South Korea.,Immunotherapy Research Center, KRIBB, Daejeon, South Korea
| | - Dae-Hyuk Kweon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Jae-Ho Cheong
- Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea. .,Serverance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.
| | - Misun Won
- Personalized Genomic Medicine Research Center, KRIBB, 125 Kwahag-ro, Yuseong-gu, Daejeon, 34141, South Korea. .,KRIBB School of Bioscience, University of Science and Technology, Daejeon, South Korea. .,R&D Center, oneCureGEN, Daejeon, South Korea.
| |
Collapse
|
13
|
Wang J, Du X, Wang X, Xiao H, Jing N, Xue W, Dong B, Gao WQ, Fang YX. Tumor-derived miR-378a-3p-containing extracellular vesicles promote osteolysis by activating the Dyrk1a/Nfatc1/Angptl2 axis for bone metastasis. Cancer Lett 2022; 526:76-90. [PMID: 34801597 DOI: 10.1016/j.canlet.2021.11.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 01/08/2023]
Abstract
Most prostate cancer (PCa)-related deaths are caused by progression to bone metastasis. Recently, the importance of extracellular vesicles (EVs) in pre-metastatic niche formation has been reported. However, whether and how tumor-derived EVs interact with bone marrow macrophages (BMMs) to release EV-delivered microRNAs to promote osteolysis and induce pre-metastatic niche formation for PCa bone metastasis remain unclear. Our in vitro and in vivo functional and mechanistic assays revealed that EV-mediated release of miR-378a-3p from tumor cells was upregulated in bone-metastatic PCa, maintaining low intracellular miR-378a-3p concentration to promote proliferation and MAOA-mediated epithelial-to-mesenchymal transition. Moreover, miR-378a-3p enrichment in tumor-derived EVs was induced by hnRNPA2B1 (a transfer chaperone) overexpression. After tumor-derived EVs were taken in by BMMs, enriched miR-378a-3p promoted osteolytic progression by inhibiting Dyrk1a to improve Nfatc1 (an osteolysis-related transcription factor) nuclear translocation, to activate the expression of downstream target gene Angptl2. As a feedback, increased Angptl2 secretion into the tumor environment promoted PCa progression. In conclusion, tumor-derived miR-378a-3p-containing EVs play a significant role in PCa bone metastasis by activating the Dyrk1a/Nfatc1/Angptl2 axis in BMMs to induce osteolytic progression, making miR-378a-3p a potential predictor of metastatic PCa. Reducing the release of miR-378a-3p-containing EVs or inhibiting the recruitment of miR-378a-3p into EVs can be a therapeutic strategy against PCa metastasis.
Collapse
Affiliation(s)
- Jialin Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xinxing Du
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiao Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Huixiang Xiao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Nan Jing
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei Xue
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Baijun Dong
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Yu-Xiang Fang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
14
|
Zarychta E, Ruszkowska-Ciastek B. Cooperation between Angiogenesis, Vasculogenesis, Chemotaxis, and Coagulation in Breast Cancer Metastases Development: Pathophysiological Point of View. Biomedicines 2022; 10:biomedicines10020300. [PMID: 35203510 PMCID: PMC8869468 DOI: 10.3390/biomedicines10020300] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Breast cancer is one of the main causes of morbidity and mortality in women. Early breast cancer has a relatively good prognosis, in contrast to metastatic disease with rather poor outcomes. Metastasis formation in distant organs is a complex process requiring cooperation of numerous cells, growth factors, cytokines, and chemokines. Tumor growth, invasion, and finally systemic spread are driven by processes of angiogenesis, vasculogenesis, chemotaxis, and coagulation. This review summarizes their role in development of distant metastases in breast cancer, as well as explains the essential processes occurring throughout these actions. Abstract With almost 2.3 million new cases and 685 thousand fatal events in 2020 alone, breast cancer remains one of the main causes of morbidity and mortality in women worldwide. Despite the increasing prevalence of the disease in recent years, the number of deaths has dropped—this is mostly the result of better diagnostic and therapeutic opportunities, allowing to recognize and treat breast cancer earlier and more efficiently. However, metastatic disease still remains a therapeutic challenge. As mechanisms of tumor spread are being explored, new drugs can be implemented in clinical practice, improving the outcomes in patients with advanced disease. Formation of metastases is a complex process, which involves activation of angiogenesis, vasculogenesis, chemotaxis, and coagulation. The actions, which occur during metastatic spread are interrelated and complementary. This review summarizes their importance and mutual connections in formation of secondary tumors in breast cancer.
Collapse
|
15
|
Takano M, Hirose N, Sumi C, Yanoshita M, Nishiyama S, Onishi A, Asakawa Y, Tanimoto K. ANGPTL2 Promotes Inflammation via Integrin α5β1 in Chondrocytes. Cartilage 2021; 13:885S-897S. [PMID: 31581797 PMCID: PMC8804837 DOI: 10.1177/1947603519878242] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Angiopoietin-like protein 2 (ANGPTL2) is a secreted molecule with numerous physiologic and pathologic functions, for example, in angiogenesis, hematopoiesis, and tumorigenesis. Although recent studies implicated ANGPTL2 in chronic inflammation in mouse peritoneal macrophages, human ligamentum flavum fibroblasts, and human retinal microvascular endothelial cells, the mechanism underlying ANGPTL2-associated inflammation in chondrocytes remains unclear. Therefore, it was investigated whether ANGPTL2 is expressed in or functions in chondrocytes. METHODS Expression of ANGPTL2 and its receptor, integrin α5β1 were examined over time in ATDC5 cells using real-time RT-PCR (reverse transcription-polymerase chain reaction) analysis. ATDC5 cells were then incubated with or without ANGPTL2 for 3 hours, and expression of the IL-1β, TNF-α, COX-2, aggrecanase (ADAMTS)-5, matrix metalloproteinase (MMP)-3, and MMP-13 genes were examined using real-time RT-PCR. Additionally, phosphorylation of ERK, JNK, p38, Akt, and NF-κB was examined by western blotting. Furthermore, it was also investigated for the effect of anti-integrin α5β1 antibody on the expression of inflammatory markers and intracellular signaling pathways. RESULTS ANGPTL2 induced the phosphorylation of all 3 MAPKs, Akt, and NF-κB and dramatically upregulated the expression of inflammation-related factor genes. Inhibiting the activation of integrin α5β1 suppressed these reactions. CONCLUSION ANGPTL2 may induce inflammatory factors by stimulating the integrin α5β1/MAPKs, Akt, and NF-κB signaling pathway.
Collapse
Affiliation(s)
- Mami Takano
- Department of Orthodontics and
Craniofacial Developmental Biology, Hiroshima University Graduate School of
Biomedical and Health Sciences, Hiroshima, Japan
| | - Naoto Hirose
- Department of Orthodontics and
Craniofacial Developmental Biology, Hiroshima University Graduate School of
Biomedical and Health Sciences, Hiroshima, Japan,Naoto Hirose, Department of Orthodontics and
Craniofacial Developmental Biology, Hiroshima University Graduate School of
Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima, 734-8551,
Japan.
| | - Chikako Sumi
- Department of Orthodontics and
Craniofacial Developmental Biology, Hiroshima University Graduate School of
Biomedical and Health Sciences, Hiroshima, Japan
| | - Makoto Yanoshita
- Department of Orthodontics and
Craniofacial Developmental Biology, Hiroshima University Graduate School of
Biomedical and Health Sciences, Hiroshima, Japan
| | - Sayuri Nishiyama
- Department of Orthodontics and
Craniofacial Developmental Biology, Hiroshima University Graduate School of
Biomedical and Health Sciences, Hiroshima, Japan
| | - Azusa Onishi
- Department of Orthodontics and
Craniofacial Developmental Biology, Hiroshima University Graduate School of
Biomedical and Health Sciences, Hiroshima, Japan
| | - Yuki Asakawa
- Department of Orthodontics and
Craniofacial Developmental Biology, Hiroshima University Graduate School of
Biomedical and Health Sciences, Hiroshima, Japan
| | - Kotaro Tanimoto
- Department of Orthodontics and
Craniofacial Developmental Biology, Hiroshima University Graduate School of
Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
16
|
Thorin-Trescases N, Labbé P, Mury P, Lambert M, Thorin E. Angptl2 is a Marker of Cellular Senescence: The Physiological and Pathophysiological Impact of Angptl2-Related Senescence. Int J Mol Sci 2021; 22:12232. [PMID: 34830112 PMCID: PMC8624568 DOI: 10.3390/ijms222212232] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 02/07/2023] Open
Abstract
Cellular senescence is a cell fate primarily induced by DNA damage, characterized by irreversible growth arrest in an attempt to stop the damage. Senescence is a cellular response to a stressor and is observed with aging, but also during wound healing and in embryogenic developmental processes. Senescent cells are metabolically active and secrete a multitude of molecules gathered in the senescence-associated secretory phenotype (SASP). The SASP includes inflammatory cytokines, chemokines, growth factors and metalloproteinases, with autocrine and paracrine activities. Among hundreds of molecules, angiopoietin-like 2 (angptl2) is an interesting, although understudied, SASP member identified in various types of senescent cells. Angptl2 is a circulatory protein, and plasma angptl2 levels increase with age and with various chronic inflammatory diseases such as cancer, atherosclerosis, diabetes, heart failure and a multitude of age-related diseases. In this review, we will examine in which context angptl2 was identified as a SASP factor, describe the experimental evidence showing that angptl2 is a marker of senescence in vitro and in vivo, and discuss the impact of angptl2-related senescence in both physiological and pathological conditions. Future work is needed to demonstrate whether the senescence marker angptl2 is a potential clinical biomarker of age-related diseases.
Collapse
Affiliation(s)
- Nathalie Thorin-Trescases
- Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada; (P.L.); (P.M.); (M.L.); (E.T.)
| | - Pauline Labbé
- Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada; (P.L.); (P.M.); (M.L.); (E.T.)
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Pauline Mury
- Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada; (P.L.); (P.M.); (M.L.); (E.T.)
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Mélanie Lambert
- Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada; (P.L.); (P.M.); (M.L.); (E.T.)
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Eric Thorin
- Montreal Heart Institute, University of Montreal, Montreal, QC H1T 1C8, Canada; (P.L.); (P.M.); (M.L.); (E.T.)
- Department of Surgery, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
17
|
Todd VM, Vecchi LA, Clements ME, Snow KP, Ontko CD, Himmel L, Pinelli C, Rafat M, Johnson RW. Hypoxia inducible factor signaling in breast tumors controls spontaneous tumor dissemination in a site-specific manner. Commun Biol 2021; 4:1122. [PMID: 34556788 PMCID: PMC8460839 DOI: 10.1038/s42003-021-02648-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023] Open
Abstract
Hypoxia is a common feature in tumors and induces signaling that promotes tumor cell survival, invasion, and metastasis, but the impact of hypoxia inducible factor (HIF) signaling in the primary tumor on dissemination to bone in particular remains unclear. To better understand the contributions of hypoxia inducible factor 1 alpha (HIF1α), HIF2α, and general HIF pathway activation in metastasis, we employ a PyMT-driven spontaneous murine mammary carcinoma model with mammary specific deletion of Hif1α, Hif2α, or von Hippel-Lindau factor (Vhl) using the Cre-lox system. Here we show that Hif1α or Hif2α deletion in the primary tumor decreases metastatic tumor burden in the bone marrow, while Vhl deletion increases bone tumor burden, as hypothesized. Unexpectedly, Hif1α deletion increases metastatic tumor burden in the lung, while deletion of Hif2α or Vhl does not affect pulmonary metastasis. Mice with Hif1α deleted tumors also exhibit reduced bone volume as measured by micro computed tomography, suggesting that disruption of the osteogenic niche may be involved in the preference for lung dissemination observed in this group. Thus, we reveal that HIF signaling in breast tumors controls tumor dissemination in a site-specific manner.
Collapse
Affiliation(s)
- Vera M Todd
- Graduate Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University, Nashville, TN, USA
| | - Lawrence A Vecchi
- Vanderbilt Center for Bone Biology, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Miranda E Clements
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Katherine P Snow
- Vanderbilt Center for Bone Biology, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Health, and Society, Vanderbilt University, Nashville, TN, USA
| | - Cayla D Ontko
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Lauren Himmel
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christopher Pinelli
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Marjan Rafat
- Vanderbilt Center for Bone Biology, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Rachelle W Johnson
- Graduate Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Center for Bone Biology, Vanderbilt University, Nashville, TN, USA.
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
18
|
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: 15] [Impact Index Per Article: 3.8] [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
|
19
|
Abstract
PURPOSE OF REVIEW Breast cancer frequently metastasizes to the bone and lung, but the ability to treat metastatic tumor cells remains a pressing clinical challenge. Histone deacetylases (HDACs) and histone acetyltransferases (HATs) have emerged as promising targets since these enzymes are aberrantly expressed in numerous cancers and regulate the expression of genes that drive tumorigenesis and metastasis. This review focuses on the abnormal expression of histone-modifying enzymes in cancers that have a high tropism for the bone and lung and explores the clinical use of histone deacetylase inhibitors for the treatment and prevention of metastasis to these sites. RECENT FINDINGS Preclinical studies have demonstrated that the role for HDACs is highly dependent on tumor type and stage of disease progression. HDAC inhibitors can induce apoptosis, senescence, cell differentiation, and tumor dormancy genes and inhibit angiogenesis, making these promising therapeutics for the treatment of metastatic disease. HDAC inhibitors are already FDA approved for hematologic malignancies and are in clinical trials with standard-of-care chemotherapies and targeted agents for several solid tumors, including cases of metastatic disease. However, these drugs can negatively impact bone homeostasis. Although HDAC inhibitors are not currently administered for the treatment of bone and lung metastatic disease, preclinical studies have shown that these drugs can reduce distant metastasis by targeting molecular factors and signaling pathways that drive tumor cell dissemination to these sites. Thus, HDAC inhibitors in combination with bone protective therapies may be beneficial in the treatment of bone metastatic cancers.
Collapse
Affiliation(s)
- Courtney M Edwards
- Graduate Program in Cancer Biology, Vanderbilt University, 2215b Garland Ave, 1165C Medical Research Building IV, Nashville, TN, 37232, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Rachelle W Johnson
- Graduate Program in Cancer Biology, Vanderbilt University, 2215b Garland Ave, 1165C Medical Research Building IV, Nashville, TN, 37232, USA.
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
20
|
Takeshita Y, Motohara T, Kadomatsu T, Doi T, Obayashi K, Oike Y, Katabuchi H, Endo M. Angiopoietin-like protein 2 decreases peritoneal metastasis of ovarian cancer cells by suppressing anoikis resistance. Biochem Biophys Res Commun 2021; 561:26-32. [PMID: 34000514 DOI: 10.1016/j.bbrc.2021.05.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022]
Abstract
Peritoneal metastasis is a common mode of spread of ovarian cancer. Despite therapeutic advances, some patients have intractable peritoneal metastasis. Therefore, in-depth characterization of the molecular mechanism of peritoneal metastasis is a key imperative. Angiopoietin-like protein 2 (ANGPTL2) is an inflammatory factor which activates NF-κB signaling and plays an important role in the pathogenesis of various inflammatory diseases including cancers, such as lung and breast cancer. In this study, we examined the role of ANGPTL2 in ovarian cancer peritoneal metastasis. We observed no difference of cell proliferation between ANGPTL2-expressing and control cells. In the mouse intraperitoneal xenograft model, formation of peritoneal metastasis by ANGPTL2-expressing cells was significantly decreased compared to control. In the in vitro analysis, the expressions of integrin α5β1, α6, and β4, but not those of αvβ3, α3, α4, and β1, were significantly decreased in ANGPTL2-expressing cells compared to control cells. ANGPTL2-expressing cells showed significantly inhibited adherence to laminin compared to control. In addition, we observed upregulation of anoikis (a form of programmed cell death occurring under an anchorage-independent condition) and significant decrease in the expression of Bcl-2 in ANGPTL2-expressing cells as compared to control cells. These results suggest that ANGPTL2 expression in ovarian cancer cells represses peritoneal metastasis by suppressing anoikis resistance.
Collapse
Affiliation(s)
- Yuko Takeshita
- Department of Molecular Genetics, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan; Department of Obstetrics and Gynecology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Takeshi Motohara
- Department of Obstetrics and Gynecology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Tomomitsu Doi
- Department of Molecular Biology, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Kunie Obayashi
- Department of Molecular Biology, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Hidetaka Katabuchi
- Department of Obstetrics and Gynecology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Motoyoshi Endo
- Department of Molecular Biology, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan.
| |
Collapse
|
21
|
Göbel A, Dell’Endice S, Jaschke N, Pählig S, Shahid A, Hofbauer LC, Rachner TD. The Role of Inflammation in Breast and Prostate Cancer Metastasis to Bone. Int J Mol Sci 2021; 22:5078. [PMID: 34064859 PMCID: PMC8151893 DOI: 10.3390/ijms22105078] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023] Open
Abstract
Tumor metastasis to bone is a common event in multiple forms of malignancy. Inflammation holds essential functions in homeostasis as a defense mechanism against infections and is a strategy to repair injured tissue and to adapt to stress conditions. However, exaggerated and/or persistent (chronic) inflammation may eventually become maladaptive and evoke diseases such as autoimmunity, diabetes, inflammatory tissue damage, fibrosis, and cancer. In fact, inflammation is now considered a hallmark of malignancy with prognostic relevance. Emerging studies have revealed a central involvement of inflammation in several steps of the metastatic cascade of bone-homing tumor cells through supporting their survival, migration, invasion, and growth. The mechanisms by which inflammation favors these steps involve activation of epithelial-to-mesenchymal transition (EMT), chemokine-mediated homing of tumor cells, local activation of osteoclastogenesis, and a positive feedback amplification of the protumorigenic inflammation loop between tumor and resident cells. In this review, we summarize established and evolving concepts of inflammation-driven tumorigenesis, with a special focus on bone metastasis.
Collapse
Affiliation(s)
- Andy Göbel
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Stefania Dell’Endice
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Nikolai Jaschke
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
- Center for Healthy Aging, Technische Universität Dresden, 01159 Dresden, Germany
| | - Sophie Pählig
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
| | - Amna Shahid
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
| | - Lorenz C. Hofbauer
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Center for Healthy Aging, Technische Universität Dresden, 01159 Dresden, Germany
| | - Tilman D. Rachner
- Mildred Scheel Early Career Center, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, 01159 Dresden, Germany; (S.D.); (N.J.); (S.P.); (A.S.); (L.C.H.); (T.D.R.)
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Center for Healthy Aging, Technische Universität Dresden, 01159 Dresden, Germany
| |
Collapse
|
22
|
Xie P, Wang H, Fang J, Du D, Tian Z, Zhen J, Liu Y, Ding Y, Fu B, Liu F, Huang D, Yu J. CSN5 Promotes Carcinogenesis of Thyroid Carcinoma Cells Through ANGPTL2. Endocrinology 2021; 162:6122687. [PMID: 33508120 DOI: 10.1210/endocr/bqaa206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Indexed: 12/13/2022]
Abstract
COP9 signalosome subunit 5 (CSN5) plays a key role in carcinogenesis of multiple cancers and contributes to the stabilization of target proteins through deubiquitylation. However, the underlying role of CSN5 in thyroid carcinoma has not been reported. In this research, our data showed that CSN5 was overexpressed in thyroid carcinoma tissues compared with paracancerous tissues. Furthermore, a series of gain/loss functional assays were performed to demonstrate the role of CSN5 in facilitating thyroid carcinoma cell proliferation and metastasis. Additionally, we found there was a positive correlation between CSN5 and angiopoietin-like protein 2 (ANGPTL2) protein levels in thyroid carcinoma tissues and that CSN5 promoted thyroid carcinoma cell proliferation and metastasis through ANGPTL2. We also identified the underlying mechanism that CSN5 elevated ANGPTL2 protein level by directly binding it, decreasing its ubiquitination and degradation. Overall, our results highlight the significance of CSN5 in promoting thyroid carcinoma carcinogenesis and implicate CSN5 as a promising candidate for thyroid carcinoma treatment.
Collapse
Affiliation(s)
- Peiyi Xie
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hui Wang
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, China National Research Center for Metabolic Diseases, Shanghai Jiao Tong University School of Medicine, China
| | - Jiayu Fang
- Second College of Clinical Medicine, Nanchang University, China
| | - Dongnian Du
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ze Tian
- Second College of Clinical Medicine, Nanchang University, China
| | - Jing Zhen
- Second College of Clinical Medicine, Nanchang University, China
| | - Yue Liu
- Second College of Clinical Medicine, Nanchang University, China
| | - Yongqi Ding
- Second College of Clinical Medicine, Nanchang University, China
| | - Bidong Fu
- Second College of Clinical Medicine, Nanchang University, China
| | - Fanrong Liu
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Da Huang
- Department of Thyroid Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jichun Yu
- Department of Thyroid Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
23
|
The Signaling Duo CXCL12 and CXCR4: Chemokine Fuel for Breast Cancer Tumorigenesis. Cancers (Basel) 2020; 12:cancers12103071. [PMID: 33096815 PMCID: PMC7590182 DOI: 10.3390/cancers12103071] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/05/2020] [Accepted: 10/18/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Breast cancer remains the most common malignancy in women. In this review, we explore the role of the CXCL12/CXCR4 pathway in breast cancer. We show that the CXCL12/CXCR4 cascade is involved in nearly every aspect of breast cancer tumorigenesis including proliferation, cell motility and distant metastasis. Moreover, we summarize current knowledge about the CXCL12/CXCR4-targeted therapies. Due to the critical roles of this pathway in breast cancer and other malignancies, we believe that audiences in different fields will find this overview helpful. Abstract The CXCL12/CXCR4 signaling pathway has emerged in the recent years as a key player in breast cancer tumorigenesis. This pathway controls many aspects of breast cancer development including cancer cell proliferation, motility and metastasis to all target organs. Moreover, the CXCL12/CXCR4 cascade affects both immune and stromal cells, creating tumor-supporting microenvironment. In this review, we examine state-of-the-art knowledge about detrimental roles of the CXCL12/CXCR4 signaling, discuss its therapeutic potential and suggest further research directions beneficial both for basic research and personalized medicine in breast cancer.
Collapse
|
24
|
Horiguchi H, Kadomatsu T, Miyata K, Terada K, Sato M, Torigoe D, Morinaga J, Moroishi T, Oike Y. Stroma-derived ANGPTL2 establishes an anti-tumor microenvironment during intestinal tumorigenesis. Oncogene 2020; 40:55-67. [PMID: 33051596 DOI: 10.1038/s41388-020-01505-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022]
Abstract
Previous studies show that tumor cell-derived angiopoietin-like protein 2 (ANGPTL2) functions as a tumor promoter in some cancer contexts. However, we recently reported that host ANGPTL2 also shows tumor suppressive activity by enhancing dendritic cell-mediated CD8+ T cell anti-tumor immune responses in mouse kidney cancer and murine syngeneic models. However, mechanisms underlying ANGPTL2-mediated tumor suppression are complex and not well known. Here, we investigated ANGPTL2 tumor suppressive function in chemically-induced intestinal tumorigenesis. ANGPTL2 deficiency enhanced intestinal tumor growth in an experimental mouse colitis-associated colon cancer (CAC) model. Angptl2-deficient mice also showed a decrease not only in CD8+ T cell responses but in CD4+ T cell responses during intestinal tumorigenesis. Furthermore, we show that stroma-derived ANGPTL2 can activate the myeloid immune response. Notably, ANGPTL2 drove generation of immunostimulatory macrophages via the NF-κB pathway, accelerating CD4+ T helper 1 (Th1) cell activation. These findings overall provide novel insight into the complex mechanisms underlying ANGPTL2 anti-tumor function in cancer pathology.
Collapse
Affiliation(s)
- Haruki Horiguchi
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan.,Department of Aging and Geriatric Medicine, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan. .,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
| | - Keishi Miyata
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Kazutoyo Terada
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Michio Sato
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan.,Institute of Resource Development and Analysis (IRDA), Kumamoto University, Kumamoto, 860-0811, Japan
| | - Daisuke Torigoe
- Institute of Resource Development and Analysis (IRDA), Kumamoto University, Kumamoto, 860-0811, Japan
| | - Jun Morinaga
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Toshiro Moroishi
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.,Department of Cell Signaling and Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Kawaguchi, 332-0012, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan. .,Department of Aging and Geriatric Medicine, Graduate School of Medical Science, Kumamoto University, Kumamoto, 860-8556, Japan. .,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
| |
Collapse
|
25
|
Wang Y, Ren S, Wang Z, Wang Z, Zhu N, Cai D, Ye Z, Ruan J. Chemokines in bone-metastatic breast cancer: Therapeutic opportunities. Int Immunopharmacol 2020; 87:106815. [PMID: 32711376 DOI: 10.1016/j.intimp.2020.106815] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022]
Abstract
Due to non-response to chemotherapy, incomplete surgical resection, and resistance to checkpoint inhibitors, breast cancer with bone metastasis is notoriously difficult to cure. Therefore, the development of novel, efficient strategies to tackle bone metastasis of breast cancer is urgently needed. Chemokines, which induce directed migration of immune cells and act as guide molecules between diverse cells and tissues, are small proteins indispensable in immunity. These complex chemokine networks play pro-tumor roles or anti-tumor roles when produced by breast cancer cells in the tumor microenvironment. Additionally, chemokines have diverse roles when secreted by various immune cells in the tumor microenvironment of breast cancer, which can be roughly divided into immunosuppressive effects and immunostimulatory effects. Recently, targeting chemokine networks has been shown to have potential for use in treatment of metastatic malignancies, including bone-metastatic breast cancer. In this review, we focus on the role of chemokines networks in the biology of breast cancer and metastasis to the bone. We also discuss the therapeutic opportunities and future prospects of targeting chemokine networks, in combination with other current standard therapies, for the treatment of bone-metastatic breast cancer.
Collapse
Affiliation(s)
| | - Shihong Ren
- First People's Hospital of Wenling, Wenling, China
| | - Zhan Wang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zenan Wang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ning Zhu
- Hebei North University, Zhangjiakou, China
| | | | - Zhaoming Ye
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | | |
Collapse
|
26
|
The Role of Adipokines and Bone Marrow Adipocytes in Breast Cancer Bone Metastasis. Int J Mol Sci 2020; 21:ijms21144967. [PMID: 32674405 PMCID: PMC7404398 DOI: 10.3390/ijms21144967] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023] Open
Abstract
The morbidity and mortality of breast cancer is mostly due to a distant metastasis, especially to the bone. Many factors may be responsible for bone metastasis in breast cancer, but interactions between tumor cells and other surrounding types of cells, and cytokines secreted by both, are expected to play the most important role. Bone marrow adipocyte (BMA) is one of the cell types comprising the bone, and adipokine is one of the cytokines secreted by both breast cancer cells and BMAs. These BMAs and adipokines are known to be responsible for cancer progression, and this review is focused on how BMAs and adipokines work in the process of breast cancer bone metastasis. Their potential as suppressive targets for bone metastasis is also explored in this review.
Collapse
|
27
|
Leblond MM, Tillé L, Nassiri S, Gilfillan CB, Imbratta C, Schmittnaegel M, Ries CH, Speiser DE, Verdeil G. CD40 Agonist Restores the Antitumor Efficacy of Anti-PD1 Therapy in Muscle-Invasive Bladder Cancer in an IFN I/II-Mediated Manner. Cancer Immunol Res 2020; 8:1180-1192. [DOI: 10.1158/2326-6066.cir-19-0826] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/24/2020] [Accepted: 07/01/2020] [Indexed: 11/16/2022]
|
28
|
Tang C, Chen E, Peng K, Wang H, Cheng X, Wang Y, Yu S, Yu Y, Cui Y, Liu T. Mining the role of angiopoietin-like protein family in gastric cancer and seeking potential therapeutic targets by integrative bioinformatics analysis. Cancer Med 2020; 9:4850-4863. [PMID: 32410376 PMCID: PMC7333835 DOI: 10.1002/cam4.3100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 11/24/2019] [Accepted: 04/16/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The indistinctive effects of antiangiogenesis agents in gastric cancer (GC) can be attributed to multifaceted gene dysregulation associated with angiogenesis. Angiopoietin-like (ANGPTL) proteins are secreted proteins regulating angiogenesis. They are also involved in inflammation and metabolism. Emerging evidences have revealed their various roles in carcinogenesis and metastasis development. However, the mRNA expression profiles, prognostic values, and biological functions of ANGPTL proteins in GC are still elucidated. METHODS We compared the transcriptional expression levels of ANGPTL proteins between GC and normal gastric tissues using ONCOMINE and TCGA-STAD. The prognostic values were evaluated by LinkedOmics and Kaplan-Meier Plotter, while the association of expression levels with clinicopathological features was generated through cBioPortal. We conducted the functional enrichment analysis with Metascape. RESULTS The expression of ANGPTL1/3/6 was lower in GC tissues than in normal gastric tissues. High expression of ANGPTL1/2/4 was correlated with short overall survival and post-progression survival in GC patients. Upregulated ANGPTL1/2 was correlated with higher histological grade, non-intestinal Lauren classification, and advanced T stage, while ANGPTL4 exhibited high expression in early T stage, M1 stage, and non-intestinal Lauren classification. CONCLUSIONS Integrative bioinformatics analysis suggests that ANGPTL1/2/4 may be potential therapeutic targets in GC patients. Among them, ANGPTL2 acts as a GC promoter, while ANGPTL1/4's role in GC is still uncertain.
Collapse
Affiliation(s)
- Cheng Tang
- Department of Medical OncologyZhongshan Hospital Affiliated to Fudan UniversityShanghaiPR China
| | - Erbao Chen
- Department of Medical OncologyZhongshan Hospital Affiliated to Fudan UniversityShanghaiPR China
| | - Ke Peng
- Department of Medical OncologyZhongshan Hospital Affiliated to Fudan UniversityShanghaiPR China
| | - Haiwei Wang
- Department of Medical OncologyZhongshan Hospital Affiliated to Fudan UniversityShanghaiPR China
| | - Xi Cheng
- Department of Medical OncologyZhongshan Hospital Affiliated to Fudan UniversityShanghaiPR China
| | - Yan Wang
- Department of Medical OncologyZhongshan Hospital Affiliated to Fudan UniversityShanghaiPR China
| | - Shan Yu
- Department of Medical OncologyZhongshan Hospital Affiliated to Fudan UniversityShanghaiPR China
| | - Yiyi Yu
- Department of Medical OncologyZhongshan Hospital Affiliated to Fudan UniversityShanghaiPR China
| | - Yuehong Cui
- Department of Medical OncologyZhongshan Hospital Affiliated to Fudan UniversityShanghaiPR China
| | - Tianshu Liu
- Department of Medical OncologyZhongshan Hospital Affiliated to Fudan UniversityShanghaiPR China
| |
Collapse
|
29
|
Singh S, Ray LA, Shahi Thakuri P, Tran S, Konopka MC, Luker GD, Tavana H. Organotypic breast tumor model elucidates dynamic remodeling of tumor microenvironment. Biomaterials 2020; 238:119853. [PMID: 32062146 PMCID: PMC8165649 DOI: 10.1016/j.biomaterials.2020.119853] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 12/13/2022]
Abstract
Fibroblasts are a critical component of tumor microenvironments and associate with cancer cells physically and biochemically during different stages of the disease. Existing cell culture models to study interactions between fibroblasts and cancer cells lack native tumor architecture or scalability. We developed a scalable organotypic model by robotically encapsulating a triple negative breast cancer (TNBC) cell spheroid within a natural extracellular matrix containing dispersed fibroblasts. We utilized an established CXCL12 - CXCR4 chemokine-receptor signaling in breast tumors to validate our model. Using imaging techniques and molecular analyses, we demonstrated that CXCL12-secreting fibroblasts have elevated activity of RhoA/ROCK/myosin light chain-2 pathway and rapidly and significantly contract collagen matrices. Signaling between TNBC cells and CXCL12-producing fibroblasts promoted matrix invasion of cancer cells by activating oncogenic mitogen-activated protein kinase signaling, whereas normal fibroblasts significantly diminished TNBC cell invasiveness. We demonstrated that disrupting CXCL12 - CXCR4 signaling using a molecular inhibitor significantly inhibited invasiveness of cancer cells, suggesting blocking of tumor-stromal interactions as a therapeutic strategy especially for cancers such as TNBC that lack targeted therapies. Our organotypic tumor model mimics native solid tumors, enables modular addition of different stromal cells and extracellular matrix proteins, and allows high throughput compound screening against tumor-stromal interactions to identify novel therapeutics.
Collapse
Affiliation(s)
- Sunil Singh
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Lucille A Ray
- Department of Chemistry, The University of Akron, Akron, OH, 44325, USA
| | - Pradip Shahi Thakuri
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Sydnie Tran
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Michael C Konopka
- Department of Chemistry, The University of Akron, Akron, OH, 44325, USA
| | - Gary D Luker
- Department of Radiology, Microbiology and Immunology, Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA.
| |
Collapse
|
30
|
Osumi H, Horiguchi H, Kadomatsu T, Tashiro K, Morinaga J, Takahashi T, Ikeda K, Ito T, Suzuki M, Endo M, Oike Y. Tumor cell-derived angiopoietin-like protein 2 establishes a preference for glycolytic metabolism in lung cancer cells. Cancer Sci 2020; 111:1241-1253. [PMID: 32012400 PMCID: PMC7156862 DOI: 10.1111/cas.14337] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 01/17/2020] [Accepted: 01/22/2020] [Indexed: 01/03/2023] Open
Abstract
We previously revealed that tumor cell‐derived angiopoietin‐like protein 2 (ANGPTL2) accelerates the metastatic capacity of tumors in an autocrine/paracrine manner by activating tumor cell motility and invasiveness and the epithelial‐mesenchymal transition. However, the effects of ANGPTL2 on cancer cell glycolytic metabolism, which is a hallmark of tumor cells, are unknown. Here we report evidence supporting a role for tumor cell‐derived ANGPTL2 in establishing a preference for glycolytic metabolism. We report that a highly metastatic lung cancer cell subline expressing abundant ANGPTL2 showed upregulated expression of the glucose transporter GLUT3 as well as enhanced glycolytic metabolism relative to a less metastatic parental line. Most notably, ANGPTL2 overexpression in the less metastatic line activated glycolytic metabolism by increasing GLUT3 expression. Moreover, ANGPTL2 signaling through integrin α5β1 increased GLUT3 expression by increasing transforming growth factor‐β (TGF‐β) signaling and expression of the downstream transcription factor zinc finger E‐box binding homeobox 1 (ZEB1). Conversely, ANGPTL2 knockdown in the highly metastatic subline decreased TGF‐β1, ZEB1, and GLUT3 expression and antagonized glycolytic metabolism. In primary tumor cells from patients with lung cancer, ANGPTL2 expression levels correlated with GLUT3 expression. Overall, this work suggests that tumor cell‐derived ANGPTL2 accelerates activities associated with glycolytic metabolism in lung cancer cells by activating TGF‐β‐ZEB1‐GLUT3 signaling.
Collapse
Affiliation(s)
- Hironobu Osumi
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.,Department of Thoracic Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Haruki Horiguchi
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.,Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kyosei Tashiro
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Jun Morinaga
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | | | - Koei Ikeda
- Department of Thoracic Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Takaaki Ito
- Department of Pathology and Experimental Medicine, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Makoto Suzuki
- Department of Thoracic Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Motoyoshi Endo
- Department of Molecular Biology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
31
|
Li Y, Su P, Wang Y, Zhang H, Liang Y, Zhang N, Song X, Li X, Li J, Yang Q. Impact of histotypes on preferential organ-specific metastasis in triple-negative breast cancer. Cancer Med 2019; 9:872-881. [PMID: 31814295 PMCID: PMC6997059 DOI: 10.1002/cam4.2759] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/13/2019] [Accepted: 11/21/2019] [Indexed: 02/06/2023] Open
Abstract
Background The distant metastasis was the most predictive characters of poor prognosis for triple‐negative breast cancer (TNBC). We aimed to evaluate the correlation between patient characters and preferential distant metastatic sites (DMS) and its effects on prognosis. Methods Using the 2010‐2014 Surveillance, Epidemiology, and End Results Program (SEER) data, patients with TNBC were classified into eight histologic subtypes. Patient characters were compared using a chi‐squared test. Logistic regression was used for identification of predictive factors. The log‐rank testing was utilized with disease‐specific survival (DSS) and overall survival (OS) as the primary outcomes. Results A total of 23 270 patients with TNBC were involved, including 1544 patients with distant metastatic cancer. Bone metastasis was diagnosed in 559 cases, brain metastasis in 124 cases, liver metastasis found in 369 cases and lung metastasis in 492 cases. Histologic subtypes including metaplastic breast carcinoma and invasive lobular carcinoma showed significant differences in preferential DMS compared with invasive ductal carcinoma. Furthermore, we found different histologic subtypes with specific DMS showed various prognosis. We also evaluated different DMS of specific histologic subtypes showed different prognosis. Conclusion Certain histologic subtypes of breast cancer are associated with preferential DMS and prognosis; this knowledge may help to further understand the mechanism of breast cancer metastasis and to monitor the prognosis of patients with TNBC.
Collapse
Affiliation(s)
- Yaming Li
- Department of Breast Surgery, Qilu Hospital, Shandong University, School of Medicine, Ji'nan, Shandong, China
| | - Peng Su
- Department of Pathology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China
| | - Yifei Wang
- Department of Breast Surgery, Qilu Hospital, Shandong University, School of Medicine, Ji'nan, Shandong, China
| | - Hanwen Zhang
- Department of Breast Surgery, Qilu Hospital, Shandong University, School of Medicine, Ji'nan, Shandong, China
| | - Yiran Liang
- Department of Breast Surgery, Qilu Hospital, Shandong University, School of Medicine, Ji'nan, Shandong, China
| | - Ning Zhang
- Department of Breast Surgery, Qilu Hospital, Shandong University, School of Medicine, Ji'nan, Shandong, China
| | - Xiaojin Song
- Department of Breast Surgery, Qilu Hospital, Shandong University, School of Medicine, Ji'nan, Shandong, China
| | - Xiaoyan Li
- Department of Breast Surgery, Qilu Hospital, Shandong University, School of Medicine, Ji'nan, Shandong, China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, China
| | - Qifeng Yang
- Department of Breast Surgery, Qilu Hospital, Shandong University, School of Medicine, Ji'nan, Shandong, China.,Pathology Tissue Bank, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
32
|
Horiguchi H, Kadomatsu T, Kurahashi R, Hara C, Miyata K, Baba M, Osumi H, Terada K, Araki K, Takai T, Kamba T, Linehan WM, Moroishi T, Oike Y. Dual functions of angiopoietin-like protein 2 signaling in tumor progression and anti-tumor immunity. Genes Dev 2019; 33:1641-1656. [PMID: 31727773 PMCID: PMC6942048 DOI: 10.1101/gad.329417.119] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/07/2019] [Indexed: 12/22/2022]
Abstract
Angiopoietin-like protein 2 (ANGPTL2) is a secreted glycoprotein homologous to angiopoietins. Previous studies suggest that tumor cell-derived ANGPTL2 has tumor-promoting function. Here, we conducted mechanistic analysis comparing ANGPTL2 function in cancer progression in a murine syngeneic model of melanoma and a mouse model of translocation renal cell carcinoma (tRCC). ANGPTL2 deficiency in tumor cells slowed tRCC progression, supporting a tumor-promoting role. However, systemic ablation of ANGPTL2 accelerated tRCC progression, supporting a tumor-suppressing role. The syngeneic model also demonstrated a tumor-suppressing role of ANGPTL2 in host tumor microenvironmental cells. Furthermore, the syngeneic model showed that PDGFRα+ fibroblasts in the tumor microenvironment express abundant ANGPTL2 and contribute to tumor suppression. Moreover, host ANGPTL2 facilitates CD8+ T-cell cross-priming and enhances anti-tumor immune responses. Importantly, ANGPTL2 activates dendritic cells through PIR-B-NOTCH signaling and enhances tumor vaccine efficacy. Our study provides strong evidence that ANGPTL2 can function in either tumor promotion or suppression, depending on what cell type it is expressed in.
Collapse
Affiliation(s)
- Haruki Horiguchi
- Department of Molecular Genetics, Graduate school of Medical science, Kumamoto University, Kumamoto 860-8556, Japan.,Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate school of Medical science, Kumamoto University, Kumamoto 860-8556, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Ryoma Kurahashi
- Department of Molecular Genetics, Graduate school of Medical science, Kumamoto University, Kumamoto 860-8556, Japan.,Department of Urology, Graduate school of Medical science, Kumamoto University, Chuo-ku, Kumamoto 860-8556, Japan
| | - Chiaki Hara
- Department of Molecular Genetics, Graduate school of Medical science, Kumamoto University, Kumamoto 860-8556, Japan.,Department of Urology, Graduate school of Medical science, Kumamoto University, Chuo-ku, Kumamoto 860-8556, Japan
| | - Keishi Miyata
- Department of Molecular Genetics, Graduate school of Medical science, Kumamoto University, Kumamoto 860-8556, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Masaya Baba
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto 860-0811, Japan
| | - Hironobu Osumi
- Department of Molecular Genetics, Graduate school of Medical science, Kumamoto University, Kumamoto 860-8556, Japan.,Department of Thoracic Surgery, Graduate school of Medical science, Kumamoto University, Kumamoto 860-8556, Japan
| | - Kazutoyo Terada
- Department of Molecular Genetics, Graduate school of Medical science, Kumamoto University, Kumamoto 860-8556, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Kimi Araki
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan.,Division of Developmental Genetics, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan
| | - Toshiyuki Takai
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Tomomi Kamba
- Department of Urology, Graduate school of Medical science, Kumamoto University, Chuo-ku, Kumamoto 860-8556, Japan
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Toshiro Moroishi
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan.,Department of Molecular Enzymology, Faculty of Life sciences, Kumamoto University, Kumamoto 860-8556, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Kawaguchi 332-0012, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate school of Medical science, Kumamoto University, Kumamoto 860-8556, Japan.,Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan.,Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Tokyo 100-0004, Japan
| |
Collapse
|
33
|
Wang X, Hu Z, Wang Z, Cui Y, Cui X. Angiopoietin-like protein 2 is an important facilitator of tumor proliferation, metastasis, angiogenesis and glycolysis in osteosarcoma. Am J Transl Res 2019; 11:6341-6355. [PMID: 31737187 PMCID: PMC6834488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 08/28/2019] [Indexed: 06/10/2023]
Abstract
Background: Solid tumors are often exposed to hypoxia. Hypoxia inducible factor (HIF-1α) upregulates numerous target genes associated with the malignant behavior of hypoxic cancer cells. Angiopoietin-like protein 2 (Angptl2), a member of the angiopoietin family, is a hypoxia-inducible gene. However, the role and potential mechanism of Angptl2, and the relationship between Angptl2 and hypoxia in osteosarcoma (OS) remain unclear. Methods: In this study, quantitative RT-PCR was performed to detect the levels of Angptl2 and HIF-1α, and western blot assay was performed to measure the expression of Angptl2, HIF-1α, CDK2, cyclin E1, P21, MMP2, MMP9, VEGFA, Ang II and HK2 in osteosarcoma cells and tissue. Subsequently, cell viability and cycle were analyzed using CCK-8 and flow cytometer assays. Cell migration, invasion and glycolysis were analyzed with Transwell, Scratch Test and glucose/lactic acid detection kits, respectively. Experiments in vivo were performed to value the effects of Angptl2 on the growth of osteosarcoma xenografts in mice. Immunofluorescent and immunohistochemistry staining were conducted to detect the expression of Ki-67 and Angptl2, respectively. Results: The results demonstrated that Angptl2 was highly expressed in OS cells, which was induced by hypoxia (HIF-1α). Additionally, Angptl2 overexpression regulated cell proliferation, invasion, migration and G1 phase arrest in OS cells. Moreover, Angptl2 promoted OS tumor growth in vivo tumor xenografts. Angptl2 might enhance angiogenesis and glycolysis by promoting VEGFA, Ang II and HK2 both in vitro and in vivo. Conclusion: In conclusion, the present findings indicated that hypoxia-induced Angptl2 expression was independent of HIF-1α in hypoxic OS cells. Angptl2 might promote OS cell proliferation, metastasis, angiogenesis and glycolysis, which could be regarded as a favorable marker for predicting a long survival time in patients with OS.
Collapse
Affiliation(s)
- Xiuhui Wang
- Department of Orthopedics, Zhoupu Hospital Affiliated to Shanghai University of Medicine and Health SciencesShanghai 201318, China
| | - Zhaohui Hu
- Department of Orthopedics, Liuzhou People’s HospitalNo. 8 Wenchang Road, Liuzhou 545006, Guangxi, China
| | - Zhe Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan UniversityShanghai 200032, China
| | - Yin Cui
- Department of Orthopedics, Zhoupu Hospital Affiliated to Shanghai University of Medicine and Health SciencesShanghai 201318, China
| | - Xu Cui
- Department of Orthopedics, Zhoupu Hospital Affiliated to Shanghai University of Medicine and Health SciencesShanghai 201318, China
| |
Collapse
|
34
|
Chen E, Tang C, Peng K, Cheng X, Wei Y, Liu T. ANGPTL6-mediated angiogenesis promotes alpha fetoprotein-producing gastric cancer progression. Pathol Res Pract 2019; 215:152454. [PMID: 31146977 DOI: 10.1016/j.prp.2019.152454] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 05/17/2019] [Indexed: 02/08/2023]
Abstract
Alpha-fetoprotein (AFP)-producing gastric cancer (AFPGC) is regarded as a rare but highly malignant gastric adenocarcinoma subtype and its clinic pathological presentation mimics hepatocellular carcinoma. However, the underlying mechanism of this disease remains elusive. The level of ANGPTL6 in AFPGC cell lines is much higher than that of common types of gastric cancer cells. A high level of ANGPTL6 confers a poor prognosis and is correlated with the expression of CD34 (an endothelial cell marker). ANGPTL6 promotes endothelial cell migration and tube formation, Moreover, ANGPTL6 knockdown inhibits cancer cell apoptosis and invasiveness. Mechanistically, ANGPTL6 activates the ERK1/2 and AKT pathways. Treatment of ERK1/2 or AKT inhibitor can attenuated cell migration and tube formation. ANGPTL6 loss results in tumor growth in vivo. Our study revealed that ANGPTL6 is an important driver gene of angiogenesis in AFPGC development. These findings provide not only an effective biomarker for diagnosis but also an attractive therapeutic target for use in AFPGC patients.
Collapse
Affiliation(s)
- Erbao Chen
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Tang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ke Peng
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xi Cheng
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yichou Wei
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China; Center of Evidence-based Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
35
|
Yang L, Sun R, Wang Y, Fu Y, Zhang Y, Zheng Z, Ji Z, Zhao D. Expression of ANGPTL2 and its impact on papillary thyroid cancer. Cancer Cell Int 2019; 19:204. [PMID: 31384179 PMCID: PMC6668118 DOI: 10.1186/s12935-019-0908-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 07/15/2019] [Indexed: 02/08/2023] Open
Abstract
Background Although the most thyroid carcinoma patients have good prognosis, around 20% of papillary thyroid carcinoma (PTC) patients have a high rate of metastasis and recurrence after routine treatment, which causes high lethality with these patients. Tumor proliferation, metastasis, and invasion are important predictors of PTC invasiveness and are key factors in cancer-related death. Angiopoietin-like 2 (ANGPTL2), a secreted protein which belongs to the angiopoietin (ANGPTL) family, was reported to be involved in the regulation of several different type of cancer cell proliferation and metastasis. However, whether ANGPTL2 plays a role in the progression of PTC, particularly in metastasis and recurrence of PTC, remains unclear. Hence, the purpose of this study was to evaluate the level of ANGPTL2 in PTC and normal thyroid, as well as para-cancerous tissue. Furthermore, the impact of ANGPTL2 on PTC cell proliferation, metastasis, recurrence and invasion was assessed to investigate the possibility whether ANGPTL2 may become a novel target for PTC therapy and cancer prognosis. Materials and methods The level of ANGPTL2 in PTC and para-cancerous tissue was assessed by immunohistochemistry. The biological effect of ANGPTL2 on thyroid cancer cell proliferation and metastasis was investigated by the Cell Counting Kit-8 (CCK8) assay, cell scratch test, and transwell assay. Correlations of ANGPTL2 expression levels with proliferation, migration, and metastasis of thyroid cancer were assessed with the TCGA data set and analyzed by gene set enrichment analysis. Receiver operating characteristic analysis was used to evaluate the utility of ANGPTL2 as a biomarker for prediction of thyroid cancer. Survival analysis was performed using the thyroid cancer database in K-M Plotter to detect correlations between survival time and ANGPTL2 levels. Results Current study revealed that: (1) ANGPTL2 was highly expressed in thyroid cancer in comparison with adjacent normal thyroid tissue; (2) ANGPTL2 expression was increased with thyroid tumor progression; (3) ANGPTL2 increased proliferation of thyroid cancer cells; (4) ANGPTL2 promoted migration and invasion of thyroid cancer cells; (5) high level of ANGPTL2 in thyroid cancer patients were significantly associated with a poor prognosis. The patients showed a higher metastasis and recurrence rate. Conclusion ANGPTL2 promoted and enhanced proliferation, metastasis, and invasion of thyroid cancer cells. ANGPTL2 may be considered as a potential biomarker for diagnosis and prognosis of thyroid cancer patients. Further evaluation needs to be done to analyze the possibility of taking ANGPTL2 as a prognostic marker and therapeutic target for papillary thyroid cancer.
Collapse
Affiliation(s)
- Longyan Yang
- 1Beijing Key Laboratory of Diabetes Prevention and Research, Department of Endocrinology, Luhe Hospital, Capital Medical University, Beijing, 101149 China
| | - Rongxin Sun
- 1Beijing Key Laboratory of Diabetes Prevention and Research, Department of Endocrinology, Luhe Hospital, Capital Medical University, Beijing, 101149 China
| | - Yan Wang
- 1Beijing Key Laboratory of Diabetes Prevention and Research, Department of Endocrinology, Luhe Hospital, Capital Medical University, Beijing, 101149 China
| | - Ying Fu
- 1Beijing Key Laboratory of Diabetes Prevention and Research, Department of Endocrinology, Luhe Hospital, Capital Medical University, Beijing, 101149 China
| | - Yuanyuan Zhang
- 1Beijing Key Laboratory of Diabetes Prevention and Research, Department of Endocrinology, Luhe Hospital, Capital Medical University, Beijing, 101149 China
| | - Zhaohui Zheng
- 1Beijing Key Laboratory of Diabetes Prevention and Research, Department of Endocrinology, Luhe Hospital, Capital Medical University, Beijing, 101149 China
| | - Zhili Ji
- 2Department of General Surgery, Luhe Hospital, Capital Medical University, Beijing, 101149 China
| | - Dong Zhao
- 1Beijing Key Laboratory of Diabetes Prevention and Research, Department of Endocrinology, Luhe Hospital, Capital Medical University, Beijing, 101149 China
| |
Collapse
|
36
|
Liu J, Zhao C, Liu B, Liu H, Wang L. Analgesia and curative effect of pamidronate disodium combined with chemotherapy on elderly patients with advanced metastatic bone cancer. Oncol Lett 2019; 18:771-775. [PMID: 31289553 PMCID: PMC6540329 DOI: 10.3892/ol.2019.10340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/06/2019] [Indexed: 11/06/2022] Open
Abstract
The curative effect and adverse reactions of pamidronate disodium in elderly patients with advanced metastatic bone cancer were evaluated. A total of 160 elderly patients with advanced metastatic bone cancer admitted to Affiliated Hospital of Nantong University from February 2012 to January 2015, were divided into the chemotherapy group (n=60) that received routine therapy and the pamidronate disodium group (n=100) that received pamidronate disodium therapy based on the chemotherapy. Pain relief, analgesic time, analgesic duration and side effects were compared between the two groups after treatment. The effect of pain relief in the pamidronate disodium group was significantly higher than that in the chemotherapy group (P<0.001). The total effective rate of the pamidronate disodium group was significantly higher than that of the chemotherapy group (P<0.001). The analgesic onset time in the pamidronate disodium group was earlier than in the chemotherapy group (P<0.001). The analgesic duration in the pamidronate disodium group was longer than that in the chemotherapy group (P<0.001). The incidence of adverse reactions and complications after treatment in the pamidronate disodium group was significantly less than that in the chemotherapy group (P<0.001). The results indicated that pamidronate disodium is effective in the treatment of elderly patients with advanced metastatic bone cancer and patients are less prone to adverse reactions, complications and pain, which is worthy of clinical application.
Collapse
Affiliation(s)
- Jiayong Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Chaoqian Zhao
- Department of Oncology, Zhuzhou Central Hospital, Zhuzhou, Hunan 412007, P.R. China
| | - Bin Liu
- Department of Oncology, The Second Hospital of Xiangya, Central South University, Changsha, Hunan 410011, P.R. China
| | - Hua Liu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Lixin Wang
- Department of Integrated TCM and Western Medicine, Shanghai Pulmonary Hospital Affiliated to Tongji University, Shanghai 200043, P.R. China
| |
Collapse
|
37
|
Abstract
Angiopoietins play important roles in angiogenesis and the maintenance of hematopoietic stem cells. Angiopoietin-like proteins (ANGPTLs) are identified as proteins structurally similar to angiopoietins, and the ANGPTL family now consists of eight members. ANGPTLs are secretary proteins, and some ANGPTLs are not only angiogenic factors but also proteins with multiple functions such as glucose metabolism, lipid metabolism, redox regulation and chronic inflammation. Chronic inflammation is one of the key factors in carcinogenesis and cancer growth, proliferation, invasion and metastasis. ANGPTL 2, 3, 4, 6 and 7 are pro-inflammatory factors and regulate cancer progression, while ANGPTL1 inhibits tumor angiogenesis and metastasis. In this review, we describe the roles of ANGPTLs in cancer progression and discuss the possibility of disturbing the progression of cancer by regulating ANGPTLs expression.
Collapse
Affiliation(s)
- Motoyoshi Endo
- Department of Molecular Biology, University of Occupational and Environmental Health, Japan
| |
Collapse
|
38
|
Chen W, Li J, Sun Z, Wu C, Ma J, Wang J, Liu S, Han X. Comparative pharmacokinetics of six coumarins in normal and breast cancer bone-metastatic mice after oral administration of Wenshen Zhuanggu Formula. JOURNAL OF ETHNOPHARMACOLOGY 2018; 224:36-44. [PMID: 29803570 DOI: 10.1016/j.jep.2018.05.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/08/2018] [Accepted: 05/23/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wenshen Zhuanggu Formula (WSZG) is a traditional Chinese medicine (TCM) prescription used in clinics for adjuvant treatment of breast cancer bone metastases in Longhua Hospital in China. WSZG has been reported to decrease the risk of bone metastases and alleviate the severity of bone lesions in a breast cancer xenograft model. AIM OF THE STUDY The present study aimed at investigating the pharmacokinetic behaviors of six coumarins in normal and breast cancer bone-metastatic mice following oral administration of WSZG extract. MATERIALS AND METHODS A bone-metastatic mouse model was established by intracardiac injection of MDA-MB-231BO breast cancer cells, and WSZG extract (1.60 g/kg) was given orally to the model and normal mice for 4 weeks. Then, the blood pharmacokinetic parameters of six bioactive components from WSZG (psoralen, isopsoralen, bergapten, xanthotoxin, osthole, and imperatorin) were analyzed by liquid chromatography tandem mass spectrometry. RESULTS There were significant differences in pharmacokinetic behaviors between normal and pathological states. Compared with normal mice, the model mice showed significantly increased AUC0-t and AUC0-∞ of the bioactive compounds (P < 0.05) and significantly decreased total blood clearance (CLZ/F) (P < 0.05). CONCLUSIONS The different pharmacokinetic behaviors might be partly ascribed to intestinal functional disorders and imbalance of gastrointestinal microbiota under the morbid state. The findings provide some valuable information to evaluate the clinical efficacy and safety of this TCM formula.
Collapse
Affiliation(s)
- Weiling Chen
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Jiajia Li
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Zhenping Sun
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Chunyu Wu
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Jiao Ma
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Jianyi Wang
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Sheng Liu
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Xianghui Han
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
39
|
ANGPTL2 deletion inhibits osteoclast generation by modulating NF-κB/MAPKs/Cyclin pathways. Biochem Biophys Res Commun 2018; 503:1471-1477. [PMID: 30031603 DOI: 10.1016/j.bbrc.2018.07.065] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 07/12/2018] [Indexed: 02/05/2023]
Abstract
Osteoclasts are multinucleated cells essential for bone-resorption. Successful repair of bone defciencies still remains a great challenge worldwide. The signaling factor angiopoietin-like protein 2 (ANGPTL2), one of eight ANGPTL proteins, functions in maintenance of tissue homeostasis partly through regulating inflammation. In the study, ANGPTL2 expression was promoted during osteoclast development and that suppressing ANGPTL2 alleviated osteoclast production regulated by macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL). The results suggested that ANGPTL2 knockdown inhibited M-CSF-caused proliferation of osteoclast precursor cells. Further, ANGPTL2 silence reduced nuclear factor of activated T cell c 1 (NFATC1) and NFATC4 expressions in M-CSF-treated cells, along with decreased Runx2, OPN and Colla1. Moreover, silencing ANGPTL2 down-regulated M-CSF-promoted expressions of pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, and chemoattractant protein-1 (CCL-2). Consistently, ANGPTL2 knockdown reduced M-CSF-enhanced activation of IKKα, IκBα and nuclear factor κB (NF-κB) and mitogen-activated protein kinases (MAPKs) (p38 MAPK, ERK1/2 MAPK and JNK MAPK). Additionally, knockdown of ANGPTL2 inhibited the induction of Cyclin D1, Cyclin D2 and Cyclin E1 due to M-CSF exposure. In vivo, we confirmed that ANGPTL2 knockout (KO) mice were protected against osteoporosis induced by ovariectomy (OVX), as proved by the improved bone loss and bone mineral density (BMD). Decreased expression of NFATCs was also observed in OVX-induced mice in the absence of ANGPTL2. Elevated release of pro-inflammatory cytokines was abrogated by ANGPTL2 knockout in femoral heads of mice with OVX operation, accompanied with a significant reduction of phosphorylated NF-κB and MAPKs signaling pathways. And down-regulated expression of Cyclin D1, Cyclin D2 and Cyclin E1 was observed in OVX-operated mice with ANGPTL2 knockout. Therefore, our study indicated that ANGPTL2 played an essential role in osteoclast generation through regulating the proliferation and inflammation of osteoclast lineage cells, providing new insights into the therapeutic strategy to alleviate bone loss.
Collapse
|
40
|
Neophytou C, Boutsikos P, Papageorgis P. Molecular Mechanisms and Emerging Therapeutic Targets of Triple-Negative Breast Cancer Metastasis. Front Oncol 2018. [PMID: 29520340 PMCID: PMC5827095 DOI: 10.3389/fonc.2018.00031] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Breast cancer represents a highly heterogeneous disease comprised by several subtypes with distinct histological features, underlying molecular etiology and clinical behaviors. It is widely accepted that triple-negative breast cancer (TNBC) is one of the most aggressive subtypes, often associated with poor patient outcome due to the development of metastases in secondary organs, such as the lungs, brain, and bone. The molecular complexity of the metastatic process in combination with the lack of effective targeted therapies for TNBC metastasis have fostered significant research efforts during the past few years to identify molecular “drivers” of this lethal cascade. In this review, the most current and important findings on TNBC metastasis, as well as its closely associated basal-like subtype, including metastasis-promoting or suppressor genes and aberrantly regulated signaling pathways at specific stages of the metastatic cascade are being discussed. Finally, the most promising therapeutic approaches and novel strategies emerging from these molecular targets that could potentially be clinically applied in the near future are being highlighted.
Collapse
Affiliation(s)
- Christiana Neophytou
- Department of Biological Sciences, School of Pure and Applied Sciences, University of Cyprus, Nicosia, Cyprus
| | | | | |
Collapse
|
41
|
Angiopoietin-Like Proteins in Angiogenesis, Inflammation and Cancer. Int J Mol Sci 2018; 19:ijms19020431. [PMID: 29389861 PMCID: PMC5855653 DOI: 10.3390/ijms19020431] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 12/27/2022] Open
Abstract
Altered expression of secreted factors by tumor cells or cells of the tumor microenvironment is a key event in cancer development and progression. In the last decade, emerging evidences supported the autocrine and paracrine activity of the members of the Angiopoietin-like (ANGPTL) protein family in angiogenesis, inflammation and in the regulation of different steps of carcinogenesis and metastasis development. Thus, ANGPTL proteins become attractive either as prognostic or predictive biomarkers, or as novel target for cancer treatment. Here, we outline the current knowledge about the functions of the ANGPTL proteins in angiogenesis, cancer progression and metastasis. Moreover, we discuss the most recent evidences sustaining their role as prognostic or predictive biomarkers for cancer therapy. Although the role of ANGPTL proteins in cancer has not been fully elucidated, increasing evidence suggest their key effects in the proliferative and invasive properties of cancer cells. Moreover, given the common overexpression of ANGPTL proteins in several aggressive solid tumors, and their role in tumor cells and cells of the tumor microenvironment, the field of research about ANGPTL proteins network may highlight new potential targets for the development of future therapeutic strategies.
Collapse
|
42
|
Wei X, Nie S, Liu H, Sun J, Liu J, Li J, Li S, Wang S, Han S, Wang J, Sun Y. Angiopoietin-like protein 2 facilitates non-small cell lung cancer progression by promoting the polarization of M2 tumor-associated macrophages. Am J Cancer Res 2017; 7:2220-2233. [PMID: 29218246 PMCID: PMC5714751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 10/03/2017] [Indexed: 06/07/2023] Open
Abstract
The functional phenotypes (M1 and M2) of tumor-associated macrophages (TAMs) are influenced by the tumor microenvironment (TME) and contribute greatly to the development of non-small cell lung cancer (NSCLC). However, the molecular mechanisms for TAM polarization remain unclear. Angiopoietin-like protein 2 (Angptl2) is involved in tumor progression. In this study, Angptl2 expression was aberrantly increased in NSCLC cells and positively correlated with TAM infiltration, tumor size and poor patient survival. Moreover, in vitro tumor cell-macrophage co-culture and recombinant protein stimulation revealed that Angptl2 fostered the M2 polarization of TAMs through the p65 nuclear factor-kappa B (NF-ĸB) pathway. In addition, Angptl2-promoted TAM enhanced proliferation, invasion, and migration of NSCLC cells and the tube formation of human umbilical vein endothelial cells (HUVECs). In vivo, TAM depletion inhibited the tumor growth induced by Angptl2. Here, for the first time, we determined that Angptl2 promoted the M2 polarization of TAMs and enhanced NSCLC progression. Interfering with Angptl2 might be an effective strategy for reprogramming TAM polarization in NSCLC, providing a promising therapy for NSCLC treatment.
Collapse
Affiliation(s)
- Xiaojuan Wei
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Siyue Nie
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Hui Liu
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Jingyu Sun
- College of Life Science, Shandong UniversityJinan, Shandong Province, China
| | - Jie Liu
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Juan Li
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Shuyan Li
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Shuyun Wang
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Shuyi Han
- Genetic and Molecular Diagnostic Center, Jinan Central Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Jun Wang
- Genetic and Molecular Diagnostic Center, Jinan Central Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| | - Yuping Sun
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan, Shandong Province, China
| |
Collapse
|
43
|
Abstract
Distant metastasis during the advanced stage of malignant tumor progression can cause considerable morbidity in cancer patients. Bone is known to be one of the most common sites of distant metastasis in patients with breast cancer (BC). BC metastases in bone are associated with excessive skeletal complications. These complications can be fatal and reduce quality of life of patients. It is important to understand the metastatic process of BC to bone to improve quality of life and design new therapeutic methods. At present, the molecular mechanisms leading to the BC metastasis to bone are not fully understood. Studying the molecular basis of BC metastasis to bone might improve our insight into this complex process. In addition, it can provide novel approaches for designing advanced and effective targeted therapies. The present article aimed to review the published papers on the molecular basis of the metastatic process of BC to bone, focusing on involved genes and signaling networks. Furthermore, we propose potential therapeutic targets that may be more effective for the inhibition and treatment of BC metastasis to bone.
Collapse
|
44
|
Torki S, Soltani A, Shirzad H, Esmaeil N, Ghatrehsamani M. Synergistic antitumor effect of NVP-BEZ235 and CAPE on MDA-MB-231 breast cancer cells. Biomed Pharmacother 2017; 92:39-45. [PMID: 28528184 DOI: 10.1016/j.biopha.2017.05.051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/07/2017] [Accepted: 05/09/2017] [Indexed: 12/11/2022] Open
Abstract
Triple negative breast cancer (TNBC) is the most lethal and aggressive kind of breast cancer. Studies with TNBC cells suggest that tumor environmental cytokines such as Transforming Growth Factor β1 (TGF-β1) have important roles in tumors fate. In the present study, we aimed to investigate, the effect of phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway dual inhibitor, NVP-BEZ235 and Caffeic acid phenyl ester (CAPE) on TNBC cell line (MDA-MB-231), stimulated with TGF-β1 for 14days in vitro. We found that TGF-β1 as a local tumor environmental cytokine plays important role in the progression and invasiveness of TNBC cells. NVP-BEZ235 inhibited the enhanced cell viability and CXCR4 expression induced by TGF-β1. In addition, the combined treatment of TNBC cell lines with CAPE and NVP-BEZ235 synergistically inhibited cell growth and reduced CXCR4 expression. Also, treatment of MDA-MB-231 cells with CAPE and NVP-BEZ235 led to decreasing the expression levels of p-FOXO3a in a time-dependent manner. Overall, these results suggest that tumor metastasis and progression in TNBC cells can be effectively reduced through the concurrent use of NVP-BEZ235 and CAPE. This could be of particular interest in assessing the effects of this therapy in the reduction of tumor metastasis and progression in other tumor types.
Collapse
Affiliation(s)
- Samira Torki
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Amin Soltani
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hedayatollah Shirzad
- Medical Plants Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdi Ghatrehsamani
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
45
|
Hu A, Huang JJ, Zhang JF, Dai WJ, Li RL, Lu ZY, Duan JL, Li JP, Chen XP, Fan JP, Xu WH, Zheng HL. Curcumin induces G2/M cell cycle arrest and apoptosis of head and neck squamous cell carcinoma in vitro and in vivo through ATM/Chk2/p53-dependent pathway. Oncotarget 2017; 8:50747-50760. [PMID: 28881600 PMCID: PMC5584201 DOI: 10.18632/oncotarget.17096] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 03/31/2017] [Indexed: 12/22/2022] Open
Abstract
Studies have demonstrated that curcumin (CUR) exerts its tumor suppressor function in a variety of human cancers including head and neck squamous cell carcinoma (HNSCC). However, the exact underlying molecular mechanisms remain obscure. Here, we aim to test whether CUR affects ATM/Chk2/p53 signaling pathway, leading to the induction of cell cycle arrest, inhibition of angiogenesis of HNSCC in vitro and in vivo. To this end, we conducted multiple methods such as MTT assay, Invasion assay, Flow cytometry, Western blotting, RT-PCR, and transfection to explore the functions and molecular insights of CUR in HNSCC. We observed that CUR significantly induced apoptosis and cell cycle arrest, inhibited angiogenesis in HNSCC. Mechanistically, we demonstrated that CUR markedly up-regulated ATM expression and subsequently down-regulated HIF-1α expression. Blockage of ATM production totally reversed CUR induced cell cycle arrest as well as anti-angiogenesis in HNSCC. Moreover, our results demonstrated that CUR exerts its antitumor activity through targeting ATM/Chk2/p53 signal pathway. In addition, the results of xenograft experiments in mice were highly consistent with in vitro studies. Collectively, our findings suggest that targeting ATM/Chk2/p53 signal pathway by CUR could be a promising therapeutic approach for HNSCC prevention and therapy.
Collapse
Affiliation(s)
- An Hu
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Shanghai, 200135, China
| | - Jing-Juan Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, 200030, China
| | - Jing-Fei Zhang
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Shanghai, 200135, China
| | - Wei-Jun Dai
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Shanghai, 200135, China
| | - Rui-Lin Li
- Department of Gerontology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Zhao-Yang Lu
- Department of Gerontology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Jun-Li Duan
- Department of Gerontology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Ji-Ping Li
- Department of Otolaryngology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Pudong New Area, Shanghai, 200127, China
| | - Xiao-Ping Chen
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Shanghai, 200135, China
| | - Jing-Ping Fan
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Shanghai, 200135, China.,Department of Otolaryngology-Head and Neck Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Wei-Hua Xu
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Shanghai, 200135, China
| | - Hong-Liang Zheng
- Department of Otolaryngology-Head and Neck Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| |
Collapse
|
46
|
Yang LK, Zhu J, Chen YH, Wang DL, Li H, Zhang LJ, Zhou JR, Liu W. Knockdown of Angiopoietin-Like Protein 2 Inhibits Proliferation and Invasion in Glioma Cells via Suppressing the ERK/MAPK Signaling Pathway. Oncol Res 2017; 25:1349-1355. [PMID: 28247845 PMCID: PMC7841190 DOI: 10.3727/096504017x14874337324615] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Angiopoietin-like protein 2 (ANGPTL2), a member of the glycoprotein family, is mainly secreted by adipose tissues under normal conditions. Recently, ANGPTL2 has been found to be upregulated in some types of cancers and is considered to be a tumor promoter. However, the functional significance of ANGPTL2 in glioma has not yet been elucidated. In this study, we investigated the specific role of ANGPTL2 in glioma. The results showed that ANGPTL2 was highly expressed in glioma tissues and cell lines. Knockdown of ANGPTL2 reduced the proliferative and invasive abilities of glioma cells. Moreover, the tumorigenesis assay showed that ANGPTL2 knockdown inhibited glioma tumor growth in vivo. We also found that ANGPTL2 knockdown decreased the protein levels of p-ERK1/2 in glioma cells and thus blocked the activity of the ERK/MAPK signaling pathway. Taken together, our study provided the first evidence that ANGPTL2 played an oncogenic role in glioma development and might be considered as a new therapeutic target for glioma treatment.
Collapse
|
47
|
Cabia B, Andrade S, Carreira MC, Casanueva FF, Crujeiras AB. A role for novel adipose tissue-secreted factors in obesity-related carcinogenesis. Obes Rev 2016; 17:361-76. [PMID: 26914773 DOI: 10.1111/obr.12377] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 12/15/2015] [Indexed: 12/12/2022]
Abstract
Obesity, a pandemic disease, is caused by an excessive accumulation of fat that can have detrimental effects on health. Adipose tissue plays a very important endocrine role, secreting different molecules that affect body physiology. In obesity, this function is altered, leading to a dysfunctional production of several factors, known as adipocytokines. This process has been linked to various comorbidities associated with obesity, such as carcinogenesis. In fact, several classical adipocytokines with increased levels in obesity have been demonstrated to exert a pro-carcinogenic role, including leptin, TNF-α, IL-6 and resistin, whereas others like adiponectin, with decreased levels in obesity, might have an anti-carcinogenic function. In this expanding field, new proteomic techniques and approaches have allowed the identification of novel adipocytokines, a number of which exhibit an altered production in obesity and type 2 diabetes and thus are related to adiposity. Many of these novel adipocytokines have also been identified in various tumour types, such as that of the breast, liver or endometrium, thereby increasing the list of potential contributors to carcinogenesis. This review is focused on the regulation of these novel adipocytokines by obesity, including apelin, endotrophin, FABP4, lipocalin 2, omentin-1, visfatin, chemerin, ANGPTL2 or osteopontin, emphasizing its involvement in tumorigenesis.
Collapse
Affiliation(s)
- B Cabia
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| | - S Andrade
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| | - M C Carreira
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| | - F F Casanueva
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| | - A B Crujeiras
- Laboratory of Molecular and Cellular Endocrinology, Instituto de Investigación Sanitaria (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS) and Santiago de Compostela University (USC), Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Madrid, Spain
| |
Collapse
|
48
|
Mills SC, Goh PH, Kudatsih J, Ncube S, Gurung R, Maxwell W, Mueller A. Cell migration towards CXCL12 in leukemic cells compared to breast cancer cells. Cell Signal 2016; 28:316-24. [DOI: 10.1016/j.cellsig.2016.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 01/19/2016] [Accepted: 01/19/2016] [Indexed: 12/23/2022]
|
49
|
Hu A, Huang JJ, Li RL, Lu ZY, Duan JL, Xu WH, Chen XP, Fan JP. Curcumin as therapeutics for the treatment of head and neck squamous cell carcinoma by activating SIRT1. Sci Rep 2015; 5:13429. [PMID: 26299580 PMCID: PMC4547100 DOI: 10.1038/srep13429] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 07/27/2015] [Indexed: 02/06/2023] Open
Abstract
SIRT1 is one of seven mammalian homologs of Sir2 that catalyzes NAD+-dependent protein deacetylation. The aim of the present study is to explore the effect of SIRT1 small molecule activator on the anticancer activity and the underlying mechanism. We examined the anticancer activity of a novel oral agent, curcumin, which is the principal active ingredient of the traditional Chinese herb Curcuma Longa. Treatment of FaDu and Cal27 cells with curcumin inhibited growth and induced apoptosis. Mechanistic studies showed that anticancer activity of curcumin is associated with decrease in migration of HNSCC and associated angiogenesis through activating of intrinsic apoptotic pathway (caspase-9) and extrinsic apoptotic pathway (caspase-8). Our data demonstrating that anticancer activity of curcumin is linked to the activation of the ATM/CHK2 pathway and the inhibition of nuclear factor-κB. Finally, increasing SIRT1 through small molecule activator curcumin has shown beneficial effects in xenograft mouse model, indicating that SIRT1 may represent an attractive therapeutic target. Our studies provide the preclinical rationale for novel therapeutics targeting SIRT1 in HNSCC.
Collapse
Affiliation(s)
- An Hu
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Miaopu Road 219, Shanghai, 200135, China
| | - Jing-Juan Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Huaihai Xi Road 241, Xuhui District, Shanghai 200030, China
| | - Rui-Lin Li
- Department of Gerontology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Kongjiang Road 1665, Shanghai, 200092, China
| | - Zhao-Yang Lu
- Department of Gerontology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Kongjiang Road 1665, Shanghai, 200092, China
| | - Jun-Li Duan
- Department of Gerontology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Kongjiang Road 1665, Shanghai, 200092, China
| | - Wei-Hua Xu
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Miaopu Road 219, Shanghai, 200135, China
| | - Xiao-Ping Chen
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Miaopu Road 219, Shanghai, 200135, China
| | - Jing-Ping Fan
- Department of Otolaryngology, Gongli Hospital, Second Military Medical University, Pudong New Area, Miaopu Road 219, Shanghai, 200135, China.,Department of Otolaryngology-Head &Neck Surgery, ChangZheng Hospital, Second Military Medical University, Fengyang Road 415, Huangpu District, Shanghai, 200003, China
| |
Collapse
|