201
|
Wu K, Zeng J, Shi X, Xie J, Li Y, Zheng H, Peng G, Zhu G, Tang D, Wu S. Targeting TIGIT Inhibits Bladder Cancer Metastasis Through Suppressing IL-32. Front Pharmacol 2021; 12:801493. [PMID: 35069212 PMCID: PMC8766971 DOI: 10.3389/fphar.2021.801493] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/24/2021] [Indexed: 02/05/2023] Open
Abstract
Bladder cancer is a highly metastatic tumor and one of the most common malignancies originating in the urinary tract. Despite the efficacy of immune checkpoints, including programmed cell death-1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), the effect of immunotherapy for bladder cancer remains unsatisfactory. Therefore, it is urgent to develop new targets to expand immunotherapeutic options. In this study, we utilized single-cell sequencing to explore the cell composition of tumors and detected a subset of Treg cells with high expression of T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) and interleukin (IL)-32. The antitumor immune response was suppressed by this subset of Treg cells, while IL-32 promoted bladder cancer metastasis. Nevertheless, targeting TIGIT not only reversed immunosuppression by restoring the antitumor immune response mediated by T cells but also suppressed the secretion of IL-32 and inhibited the metastasis of bladder cancer cells. Thus, our study provided novel insights into immunosuppression in bladder cancer and highlighted TIGIT as a novel target for immunotherapy of bladder cancer. We also illustrated the mechanism of the dual effect of targeting TIGIT and revealed the metastasis-promoting effect of IL-32 in bladder cancer. Collectively, these findings raise the possibility of utilizing TIGIT as a target against bladder cancer from the bench to the bedside.
Collapse
Affiliation(s)
- Kang Wu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Jun Zeng
- Department of Genetics and Cell Biology, College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Xulian Shi
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Jiajia Xie
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Yuqing Li
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Haoxiang Zheng
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Guoyu Peng
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Guanghui Zhu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Dongdong Tang
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Shenzhen Following Precision Medicine Research Institute, Shenzhen, China
| | - Song Wu
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, China
- Medical Laboratory of Shenzhen Luohu People’s Hospital, Shenzhen, China
- Teaching Center of Shenzhen Luohu Hospital, Shantou University Medical College, Shantou, China
- Department of Urology and Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Song Wu,
| |
Collapse
|
202
|
Li SJ, Wei XH, Zhan XM, He JY, Zeng YQ, Tian XM, Yuan ST, Sun L. Adipocyte-Derived Leptin Promotes PAI-1 -Mediated Breast Cancer Metastasis in a STAT3/miR-34a Dependent Manner. Cancers (Basel) 2020; 12:cancers12123864. [PMID: 33371368 PMCID: PMC7767398 DOI: 10.3390/cancers12123864] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 12/16/2020] [Accepted: 12/18/2020] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Although adipocytes affect the metastatic behavior of cancer cells, the underlying molecular mechanisms remain largely elusive. Thereby, we sought to screen for the signaling pathways responsible for adipocyte-induced motility of breast cancer cells by employing a breast cancer cell/adipocyte coculture system. Our study revealed that adipocyte coculture stimulated PAI-1 expression in breast cancer cells to potentiate cell motility. Furthermore, we obtained evidence that adipocytes secreted leptin to activate OBR in breast cancer cells, which phosphorylated STAT3 to promote the transcription of PAI-1 and repress the expression of miR-34a as the negative regulator of PAI-1. Our study provides new evidence for the involvement of adipocytes in breast cancer evolution, which advances the evolving roles of stromal cells in tumor pathogenesis. Abstract The crosstalk between cancer cells and adipocytes is critical for breast cancer progression. However, the molecular mechanisms underlying these interactions have not been fully characterized. In the present study, plasminogen activator inhibitor-1 (PAI-1) was found to be a critical effector of the metastatic behavior of breast cancer cells upon adipocyte coculture. Loss-of-function studies indicated that silencing PAI-1 suppressed cancer cell migration. Furthermore, we found that PAI-1 was closely related to the epithelial-mesenchymal transition (EMT) process in breast cancer patients. A loss-of-function study and a mammary orthotopic implantation metastasis model showed that PAI-1 promoted breast cancer metastasis by affecting the EMT process. In addition, we revealed that leptin/OBR mediated the regulation of PAI-1 through the interactions between adipocytes and breast cancer cells. Mechanistically, we elucidated that leptin/OBR further activated STAT3 to promote PAI-1 expression via miR-34a–dependent and miR-34a–independent mechanisms in breast cancer cells. In conclusion, our study suggests that targeting PAI-1 and interfering with its upstream regulators may benefit breast cancer patients.
Collapse
Affiliation(s)
- Si-Jing Li
- New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, China; (S.-J.L.); (X.-M.Z.); (J.-Y.H.); (Y.-Q.Z.); (X.-M.T.)
| | - Xiao-Hui Wei
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China;
| | - Xiao-Man Zhan
- New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, China; (S.-J.L.); (X.-M.Z.); (J.-Y.H.); (Y.-Q.Z.); (X.-M.T.)
| | - Jin-Yong He
- New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, China; (S.-J.L.); (X.-M.Z.); (J.-Y.H.); (Y.-Q.Z.); (X.-M.T.)
- China Cell-gene Therapy Translational Medicine Research Center, Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Yu-Qi Zeng
- New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, China; (S.-J.L.); (X.-M.Z.); (J.-Y.H.); (Y.-Q.Z.); (X.-M.T.)
| | - Xue-Mei Tian
- New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, China; (S.-J.L.); (X.-M.Z.); (J.-Y.H.); (Y.-Q.Z.); (X.-M.T.)
| | - Sheng-Tao Yuan
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (S.-T.Y.); (L.S.)
| | - Li Sun
- New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, China; (S.-J.L.); (X.-M.Z.); (J.-Y.H.); (Y.-Q.Z.); (X.-M.T.)
- Correspondence: (S.-T.Y.); (L.S.)
| |
Collapse
|
203
|
Zarrilli G, Businello G, Dieci MV, Paccagnella S, Carraro V, Cappellesso R, Miglietta F, Griguolo G, Guarneri V, Lo Mele M, Fassan M. The Tumor Microenvironment of Primitive and Metastatic Breast Cancer: Implications for Novel Therapeutic Strategies. Int J Mol Sci 2020; 21:8102. [PMID: 33143050 PMCID: PMC7662409 DOI: 10.3390/ijms21218102] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/24/2022] Open
Abstract
Breast cancer evolves thanks to a dense and close interaction with the surrounding tumor microenvironment (TME). Fibroblasts, leukocytes, blood and lymphatic endothelial cells and extracellular matrix are the constituents of this entity, and they synergistically play a pivotal role in all of the stages of breast cancer development, from its onset to its metastatic spread. Moreover, it has been widely demonstrated that variations to the TME can correspond to prognosis variations. Breast cancer not only modulates the transformation of the environment within the mammary gland, but the same process is observed in metastases as well. In this minireview, we describe the features of TME within the primitive breast cancer, throughout its evolution and spread into the main metastatic sites.
Collapse
Affiliation(s)
- Giovanni Zarrilli
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy; (G.Z.); (G.B.); (S.P.); (V.C.); (R.C.)
| | - Gianluca Businello
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy; (G.Z.); (G.B.); (S.P.); (V.C.); (R.C.)
| | - Maria Vittoria Dieci
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128 Padua, Italy; (M.V.D.); (G.G.); (V.G.)
- Department of Surgery, Oncology and Gastroenterology (DISCoG), University of Padua, 35121 Padua, Italy;
| | - Silvia Paccagnella
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy; (G.Z.); (G.B.); (S.P.); (V.C.); (R.C.)
| | - Valentina Carraro
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy; (G.Z.); (G.B.); (S.P.); (V.C.); (R.C.)
| | - Rocco Cappellesso
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy; (G.Z.); (G.B.); (S.P.); (V.C.); (R.C.)
| | - Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology (DISCoG), University of Padua, 35121 Padua, Italy;
| | - Gaia Griguolo
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128 Padua, Italy; (M.V.D.); (G.G.); (V.G.)
- Department of Surgery, Oncology and Gastroenterology (DISCoG), University of Padua, 35121 Padua, Italy;
| | - Valentina Guarneri
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128 Padua, Italy; (M.V.D.); (G.G.); (V.G.)
- Department of Surgery, Oncology and Gastroenterology (DISCoG), University of Padua, 35121 Padua, Italy;
| | - Marcello Lo Mele
- Surgical Pathology Unit, University Hospital of Padua, 35121 Padua, Italy;
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy; (G.Z.); (G.B.); (S.P.); (V.C.); (R.C.)
| |
Collapse
|
204
|
Louault K, Li RR, DeClerck YA. Cancer-Associated Fibroblasts: Understanding Their Heterogeneity. Cancers (Basel) 2020; 12:E3108. [PMID: 33114328 PMCID: PMC7690906 DOI: 10.3390/cancers12113108] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/12/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment (TME) plays a critical role in tumor progression. Among its multiple components are cancer-associated fibroblasts (CAFs) that are the main suppliers of extracellular matrix molecules and important contributors to inflammation. As a source of growth factors, cytokines, chemokines and other regulatory molecules, they participate in cancer progression, metastasis, angiogenesis, immune cell reprogramming and therapeutic resistance. Nevertheless, their role is not fully understood, and is sometimes controversial due to their heterogeneity. CAFs are heterogeneous in their origin, phenotype, function and presence within tumors. As a result, strategies to target CAFs in cancer therapy have been hampered by the difficulties in better defining the various populations of CAFs and by the lack of clear recognition of their specific function in cancer progression. This review discusses how a greater understanding of the heterogeneous nature of CAFs could lead to better approaches aimed at their use or at their targeting in the treatment of cancer.
Collapse
Affiliation(s)
- Kévin Louault
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90027, USA
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Rong-Rong Li
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA;
| | - Yves A. DeClerck
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90027, USA
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Biochemistry and Molecular Biology, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
205
|
Ortiz-Otero N, Marshall JR, Lash B, King MR. Chemotherapy-induced release of circulating-tumor cells into the bloodstream in collective migration units with cancer-associated fibroblasts in metastatic cancer patients. BMC Cancer 2020; 20:873. [PMID: 32917154 PMCID: PMC7488506 DOI: 10.1186/s12885-020-07376-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 09/02/2020] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Recent studies have shown that chemotherapy destabilizes the blood vasculature and increases circulating tumor cell (CTC) influx into the circulation of metastatic cancer patients (Met-pa). CTCs are a precursor of cancer metastasis, in which they can migrate as single CTCs or as CTC clusters with stromal cells such as cancer-associated fibroblasts (CAFs) as cell aggregates. METHODS Blood samples were collected from 52 Met-pa, and the number of CTC and CAF was determined along with the temporal fluctuation of these through the chemotherapy treatment. RESULTS In this study, CTC level was found to increase two-fold from the initial level after 1 cycle of chemotherapy and returned to baseline after 2 cycles of chemotherapy. Importantly, we determined for the first time that circulating CAF levels correlate with worse prognosis and a lower probability of survival in Met-pa. Based on the CTC release induced by chemotherapy, we evaluated the efficacy of our previously developed cancer immunotherapy to eradicate CTCs from Met-pa blood using an ex vivo approach and demonstrate this could kill over 60% of CTCs. CONCLUSION Collectively, we found that CAF levels in Met-pa serve as a predictive biomarker for cancer prognosis. Additionally, we demonstrate the efficacy of our therapy to kill primary CTCs for a range of cancer types, supporting its potential use as an anti-metastasis therapy in the clinical setting.
Collapse
Affiliation(s)
- Nerymar Ortiz-Otero
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Jocelyn R Marshall
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Bradley Lash
- Guthrie Clinical Research Center, Sayre, PA, 18840, USA
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
206
|
Su CY, Li JQ, Zhang LL, Wang H, Wang FH, Tao YW, Wang YQ, Guo QR, Li JJ, Liu Y, Yan YY, Zhang JY. The Biological Functions and Clinical Applications of Integrins in Cancers. Front Pharmacol 2020; 11:579068. [PMID: 33041823 PMCID: PMC7522798 DOI: 10.3389/fphar.2020.579068] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
Integrins are the adhesion molecules and receptors of extracellular matrix (ECM). They mediate the interactions between cells-cells and cells-ECM. The crosstalk between cancer cells and their microenvironment triggers a variety of critical signaling cues and promotes the malignant phenotype of cancer. As a type of transmembrane protein, integrin-mediated cell adhesion is essential in regulating various biological functions of cancer cells. Recent evidence has shown that integrins present on tumor cells or tumor-associated stromal cells are involved in ECM remodeling, and as mechanotransducers sensing changes in the biophysical properties of the ECM, which contribute to cancer metastasis, stemness and drug resistance. In this review, we outline the mechanism of integrin-mediated effects on biological changes of cancers and highlight the current status of clinical treatments by targeting integrins.
Collapse
Affiliation(s)
- Chao-yue Su
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jing-quan Li
- The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Ling-ling Zhang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Hui Wang
- Guangzhou Institute of Pediatrics/Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Feng-hua Wang
- Guangzhou Institute of Pediatrics/Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yi-wen Tao
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yu-qing Wang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Qiao-ru Guo
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jia-jun Li
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yun Liu
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yan-yan Yan
- Institute of Immunology and School of Medicine, Shanxi Datong University, Datong, China
| | - Jian-ye Zhang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital, Hainan Medical University, Haikou, China
| |
Collapse
|
207
|
Aass KR, Kastnes MH, Standal T. Molecular interactions and functions of IL-32. J Leukoc Biol 2020; 109:143-159. [PMID: 32869391 DOI: 10.1002/jlb.3mr0620-550r] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/29/2020] [Accepted: 07/28/2020] [Indexed: 12/16/2022] Open
Abstract
IL-32 is a multifaceted cytokine associated with several diseases and inflammatory conditions. Its expression is induced in response to cellular stress such as hypoxia, infections, and pro-inflammatory cytokines. IL-32 can be secreted from cells and can induce the production of pro-inflammatory cytokines from several cell types but are also described to have anti-inflammatory functions. The intracellular form of IL-32 is shown to play an important role in various cellular processes, including the defense against intracellular bacteria and viruses and in modulation of cell metabolism. In this review, we discuss current literature on molecular interactions of IL-32 with other proteins. We also review data on the role of intracellular IL-32 as a metabolic regulator and its role in antimicrobial host defense.
Collapse
Affiliation(s)
- Kristin Roseth Aass
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research (CEMIR), Trondheim, Norway
| | - Martin H Kastnes
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research (CEMIR), Trondheim, Norway
| | - Therese Standal
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research (CEMIR), Trondheim, Norway.,Department of Hematology, St. Olavs Hospital, Trondheim, Norway
| |
Collapse
|
208
|
Saini H, Rahmani Eliato K, Veldhuizen J, Zare A, Allam M, Silva C, Kratz A, Truong D, Mouneimne G, LaBaer J, Ros R, Nikkhah M. The role of tumor-stroma interactions on desmoplasia and tumorigenicity within a microengineered 3D platform. Biomaterials 2020; 247:119975. [PMID: 32278213 DOI: 10.1016/j.biomaterials.2020.119975] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/11/2020] [Accepted: 03/14/2020] [Indexed: 12/14/2022]
Abstract
The tumor microenvironment has been demonstrated to play a crucial role in modulating cancer progression. Amongst various cell types within the tumor microenvironment, cancer associated fibroblasts (CAFs) are in abundance, serving to modulate the biophysical properties of the stromal matrix, through excessive deposition of extracellular matrix (ECM) proteins that leads to enhanced tumor progression. There is still a critical need to develop a fundamental framework on the role of tumor-stromal cell interactions on desmoplasia and tumorigenicity. Herein, we developed a 3D microengineered organotypic tumor-stroma model incorporated with breast cancer cells surrounded by CAF-embedded collagen matrix. We further integrated our platform with atomic force microscopy (AFM) to study the dynamic changes in stromal stiffness during active tumor invasion. Our findings primarily demonstrated enhanced tumor progression in the presence of CAFs. Furthermore, we highlighted the crucial role of crosstalk between tumor cells and CAFs on stromal desmoplasia, where we identified the role of tumor-secreted PDGF-AA/-BB on elevated matrix stiffness. Inhibition of the activity of PDGFRs in CAFs led to attenuation of stromal stiffness. Overall, our work presents a well-controlled tumor microenvironment model capable of dissecting specific biophysical and biochemical signaling cues which lead to stromal desmoplasia and tumor progression.
Collapse
|
209
|
Avagliano A, Fiume G, Ruocco MR, Martucci N, Vecchio E, Insabato L, Russo D, Accurso A, Masone S, Montagnani S, Arcucci A. Influence of Fibroblasts on Mammary Gland Development, Breast Cancer Microenvironment Remodeling, and Cancer Cell Dissemination. Cancers (Basel) 2020; 12:E1697. [PMID: 32604738 PMCID: PMC7352995 DOI: 10.3390/cancers12061697] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
The stromal microenvironment regulates mammary gland development and tumorigenesis. In normal mammary glands, the stromal microenvironment encompasses the ducts and contains fibroblasts, the main regulators of branching morphogenesis. Understanding the way fibroblast signaling pathways regulate mammary gland development may offer insights into the mechanisms of breast cancer (BC) biology. In fact, the unregulated mammary fibroblast signaling pathways, associated with alterations in extracellular matrix (ECM) remodeling and branching morphogenesis, drive breast cancer microenvironment (BCM) remodeling and cancer growth. The BCM comprises a very heterogeneous tissue containing non-cancer stromal cells, namely, breast cancer-associated fibroblasts (BCAFs), which represent most of the tumor mass. Moreover, the different components of the BCM highly interact with cancer cells, thereby generating a tightly intertwined network. In particular, BC cells activate recruited normal fibroblasts in BCAFs, which, in turn, promote BCM remodeling and metastasis. Thus, comparing the roles of normal fibroblasts and BCAFs in the physiological and metastatic processes, could provide a deeper understanding of the signaling pathways regulating BC dissemination. Here, we review the latest literature describing the structure of the mammary gland and the BCM and summarize the influence of epithelial-mesenchymal transition (EpMT) and autophagy in BC dissemination. Finally, we discuss the roles of fibroblasts and BCAFs in mammary gland development and BCM remodeling, respectively.
Collapse
Affiliation(s)
- Angelica Avagliano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (G.F.); (E.V.)
| | - Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy;
| | - Nunzia Martucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Eleonora Vecchio
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (G.F.); (E.V.)
| | - Luigi Insabato
- Anatomic Pathology Unit, Department of Advanced Biomedical Sciences, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (L.I.); (D.R.)
| | - Daniela Russo
- Anatomic Pathology Unit, Department of Advanced Biomedical Sciences, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (L.I.); (D.R.)
| | - Antonello Accurso
- Department of General, Oncological, Bariatric and Endocrine-Metabolic Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Stefania Masone
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Stefania Montagnani
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| |
Collapse
|
210
|
Molecular mechanism of gossypol mediating CCL2 and IL‑8 attenuation in triple‑negative breast cancer cells. Mol Med Rep 2020; 22:1213-1226. [PMID: 32627003 PMCID: PMC7339712 DOI: 10.3892/mmr.2020.11240] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 04/28/2020] [Indexed: 12/28/2022] Open
Abstract
Chronic inflammation associated with cancer is characterized by the production of different types of chemokines and cytokines. In cancer, numerous signaling pathways upregulate the expression levels of several cytokines and evolve cells to the neoplastic state. Therefore, targeting these signaling pathways through the inhibition of distinctive gene expression is a primary target for cancer therapy. The present study investigated the anticancer effects of the natural polyphenol gossypol (GOSS) in triple-negative breast cancer (TNBC) cells, the most aggressive breast cancer type with poor prognosis. GOSS effects were examined in two TNBC cell lines: MDA-MB-231 (MM-231) and MDA-MB-468 (MM-468), representing Caucasian Americans (CA) and African Americans (AA), respectively. The obtained IC50s revealed no significant difference between the two cell lines' response to the compound. However, the use of microarray assays for cytokine determination indicated the ability of GOSS to attenuate the expression levels of cancer-related cytokines in the two cell lines. Although GOSS did not alter CCL2 expression in MM-468 cells, it was able to cause 30% inhibition in TNF-α-stimulated MM-231 cells. Additionally, IL-8 was not altered by GOSS treatment in MM-231 cells, while its expression was inhibited by 60% in TNF-α-activated MM-468 cells. ELISA assays supported the microarray data and indicated that CCL2 expression was inhibited by 40% in MM-231 cells, and IL-8 expression was inhibited by 50% in MM-468 cells. Furthermore, in MM-231 cells, GOSS inhibited CCL2 release via the repression of IKBKE, CCL2 and MAPK1 gene expression. Additionally, in MM-468 cells, the compound downregulated the release of IL-8 through repressing IL-8, MAPK1, MAPK3, CCDC88A, STAT3 and PIK3CD gene expression. In conclusion, the data obtained in the present study indicate that the polyphenol compound GOSS may provide a valuable tool in TNBC therapy.
Collapse
|
211
|
Tong Y, Yang L, Yu C, Zhu W, Zhou X, Xiong Y, Wang W, Ji F, He D, Cao X. Tumor-Secreted Exosomal lncRNA POU3F3 Promotes Cisplatin Resistance in ESCC by Inducing Fibroblast Differentiation into CAFs. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:1-13. [PMID: 32637576 PMCID: PMC7321817 DOI: 10.1016/j.omto.2020.05.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 05/27/2020] [Indexed: 12/30/2022]
Abstract
Cancer-associated fibroblasts (CAFs), an activated subpopulation of fibroblasts, occupy a central position in the tumor microenvironment and have been shown to promote chemoresistance in multiple cancer types by secreting inflammatory cytokines. Herein, we report that tumor-secreted exosomal long non-coding RNAs (lncRNAs) can regulate cisplatin resistance in esophageal squamous cell carcinoma (ESCC) through transformation of normal fibroblasts (NFs) to CAFs. Primary CAFs and matched NFs were isolated from tumor tissues and matched normal esophageal epithelial tissues of ESCC patients. Fluorescence microscopy and qRT-PCR were used to investigate the transportation of exosomal lncRNAs from ESCC cells to NFs. To identify the specific lncRNAs involved, 14 ESCC-related lncRNAs were measured in NFs after incubation with exosomes from ESCC cells. We demonstrated that lncRNA POU3F3 can be transferred from ESCC cells to NFs via exosomes and that it mediated fibroblast activation. Activated fibroblasts further promoted proliferation and cisplatin resistance of ESCC cells through secreting interleukin 6 (IL-6). Moreover, our clinical data showed that high levels of plasma exosomal lncRNA POU3F3 correlated significantly with lack of complete response and poor survival in ESCC patients. Therefore, these data demonstrate that lncRNA POU3F3 is involved in cisplatin resistance in ESCC and that this effect is mediated through exosomal lncRNA POU3F3-induced transformation of NFs to CAFs.
Collapse
Affiliation(s)
- Yusuo Tong
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Lili Yang
- Department of Oncology Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Changhua Yu
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Weiguo Zhu
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xilei Zhou
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Yaozu Xiong
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Wanwei Wang
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Fuzhi Ji
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Dongcheng He
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xiufeng Cao
- Department of Oncology Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Thoracic Surgery, Taikang Xianlin Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
212
|
Ojalill M, Virtanen N, Rappu P, Siljamäki E, Taimen P, Heino J. Interaction between prostate cancer cells and prostate fibroblasts promotes accumulation and proteolytic processing of basement membrane proteins. Prostate 2020; 80:715-726. [PMID: 32364250 DOI: 10.1002/pros.23985] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/03/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Tumor microenvironment or stroma has the potency to regulate the behavior of malignant cells. Fibroblast-like cells are abundant in tumor stroma and they are also responsible for the synthesis of many extracellular matrix components. Fibroblast-cancer cell interplay can modify the functions of both cell types. METHODS We applied mass spectrometry and proteomics to unveil the matrisome in 3D spheroids formed by DU145 prostate cancer cells, PC3 prostate cancer cells, or prostate-derived fibroblasts. Similarly, DU145/fibroblast and PC3/fibroblast coculture spheroids were also analyzed. Western blot analysis and immunofluorescence were used to confirm the presence of specific proteins in spheroids. Cancer dissemination was studied by utilizing "out of spheroids" migration and invasion assays. RESULTS In the spheroid model cancer cell-fibroblast interplay caused remarkable changes in the extracellular matrix and accelerated the invasion of DU145 cells. Fibroblasts produced structural matrix proteins, growth factors, and matrix metalloproteinases. In cancer cell/fibroblast cocultures basement membrane components, including laminins (α3, α5, β2, and β3), heparan sulfate proteoglycan (HSPG2 gene product), and collagen XVIII accumulated in a prominent manner when compared with spheroids that contained fibroblasts or cancer cells only. Furthermore, collagen XVIII was intensively processed to different endostatin-containing isoforms by cancer cell-derived cathepsin L. CONCLUSIONS Fibroblasts can promote carcinoma cell dissemination by several different mechanisms. Extracellular matrix and basement membrane proteins provide attachment sites for cell locomotion promoting adhesion receptors. Growth factors and metalloproteinases are known to accelerate cell invasion. In addition, cancer cell-fibroblast interplay generates biologically active fragments of basement membrane proteins, such as endostatin.
Collapse
Affiliation(s)
| | - Noora Virtanen
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Pekka Rappu
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Elina Siljamäki
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Pekka Taimen
- Department of Pathology, Turku University Hospital, University of Turku, Turku, Finland
| | - Jyrki Heino
- Department of Biochemistry, University of Turku, Turku, Finland
| |
Collapse
|
213
|
Salimifard S, Masjedi A, Hojjat-Farsangi M, Ghalamfarsa G, Irandoust M, Azizi G, Mohammadi H, Keramati MR, Jadidi-Niaragh F. Cancer associated fibroblasts as novel promising therapeutic targets in breast cancer. Pathol Res Pract 2020; 216:152915. [PMID: 32146002 DOI: 10.1016/j.prp.2020.152915] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/25/2020] [Accepted: 03/01/2020] [Indexed: 12/12/2022]
Abstract
Breast cancer is one of the most important women-related malignancies, which is incurable (particularly in advanced stages) and tumor microenvironment is a number one accused part in the inefficiency of current anti-breast cancer therapeutic strategies. The tumor microenvironment is composed of various cellular and acellular components, which provide an optimum condition for freely expanding cancer cells in various cancer types, particularly breast cancer. Cancer-associated fibroblasts (CAFs) are one of the main cell types in the breast tumor region, which can promote various tumor-promoting processes such as expansion, angiogenesis, metastasis and drug resistance. CAFs directly (by cell-to-cell communication) and indirectly (through secreting soluble factors) can exert their tumorigenic functions. We try to elucidate the immunobiology of CAFs, their origin, function, and heterogeneity in association with their role in various cancer-promoting processes in breast cancer. Based on current knowledge, we believe that the origin of CAFs, their subsets, and their specific expressed biomarkers determine their pro- or anti-tumor functions. Therefore, targeting CAF without considering their specific functions may lead to a deleterious outcome. We propose to find and characterize each subtype of CAFs in association with its specific function in different stages of breast cancer to develop novel promising therapeutic approaches against the right CAF subtype.
Collapse
Affiliation(s)
- Sevda Salimifard
- Department of Hematology and Blood Transfusion, School of Allied Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Masjedi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; The Persian Gulf Marine Biotechnology Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mahzad Irandoust
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gholamreza Azizi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Hamed Mohammadi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Reza Keramati
- Department of Hematology and Blood Transfusion, School of Allied Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
214
|
Wei L, Zhou Q, Tian H, Su Y, Fu GH, Sun T. Integrin β3 promotes cardiomyocyte proliferation and attenuates hypoxia-induced apoptosis via regulating the PTEN/Akt/mTOR and ERK1/2 pathways. Int J Biol Sci 2020; 16:644-654. [PMID: 32025212 PMCID: PMC6990915 DOI: 10.7150/ijbs.39414] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022] Open
Abstract
Objective: Integrin β3 is one of the main integrin heterodimer receptors on the surface of cardiac myocytes. Our previous studies showed that hypoxia induces apoptosis and increases integrin β3 expression in cardiomyocytes. However, the exact mechanism by which integrin β3 protects against apoptosis remains unclear. Hence, the present investigation aimed to explore the mechanism of integrin β3 in cardiomyocyte proliferation and hypoxia-induced cardiomyocyte apoptosis. Methods: Stable cells and in vivo acute and chronic heart failure rat models were generated to reveal the essential role of integrin β3 in cardiomyocyte proliferation and apoptosis. Western blotting and immunohistochemistry were employed to detect the expression of integrin β3 in the stable cells and rat cardiac tissue. Flow cytometer was used to investigate the role of integrin β3 in hypoxia-induced cardiomyocyte apoptosis. Confocal microscopy was used to detect the localization of integrin β3 and integrin αv in cardiomyocytes. Results: A cobaltous chloride-induced hypoxic microenvironment stimulated cardiomyocyte apoptosis and increased integrin β3 expression in H9C2 cells, AC16 cells, and cardiac tissue from acute and chronic heart failure rats. The overexpression of integrin β3 promoted cardiomyocyte proliferation, whereas silencing integrin β3 expression resulted in decreased cell proliferation in vitro. Furthermore, knocking down integrin β3 expression using shRNA or the integrin β3 inhibitor cilengitide exacerbated cobaltous chloride-induced cardiomyocyte apoptosis, whereas overexpression of integrin β3 weakened cobaltous chloride-induced cardiomyocytes apoptosis. We found that integrin β3 promoted cardiomyocytes proliferation through the regulation of the PTEN/Akt/mTOR and ERK1/2 signaling pathways. In addition, we found that knockdown of integrin αv or integrin β1 weakened the effect of integrin β3 in cardiomyocyte proliferation. Conclusion: Our findings revealed the molecular mechanism of the role of integrin β3 in cardiomyocyte proliferation and hypoxia-induced cardiomyocyte apoptosis, providing new insights into the mechanisms underlying myocardial protection.
Collapse
Affiliation(s)
- Lijiang Wei
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine. Shanghai, 200025, China
| | - Qingqing Zhou
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 20032, China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 20032, China
| | - Yifan Su
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine. Shanghai, 200025, China
| | - Guo-Hui Fu
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, No.280, South Chong-Qing Road, Shanghai 200025, People's Republic of China
| | - Ting Sun
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine. Shanghai, 200025, China
| |
Collapse
|
215
|
Byun WS, Kim WK, Han HJ, Chung HJ, Jang K, Kim HS, Kim S, Kim D, Bae ES, Park S, Lee J, Park HG, Lee SK. Targeting Histone Methyltransferase DOT1L by a Novel Psammaplin A Analog Inhibits Growth and Metastasis of Triple-Negative Breast Cancer. Mol Ther Oncolytics 2019; 15:140-152. [PMID: 31720371 PMCID: PMC6838941 DOI: 10.1016/j.omto.2019.09.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/19/2019] [Indexed: 01/24/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most intractable cancer in women with a high risk of metastasis. While hyper-methylation of histone H3 catalyzed by disruptor of telomeric silencing 1-like (DOT1L), a specific methyltransferase for histone H3 at lysine residue 79 (H3K79), is reported as a potential target for TNBCs, early developed nucleoside-type DOT1L inhibitors are not sufficient for effective inhibition of growth and metastasis of TNBC cells. We found that TNBC cells had a high expression level of DOT1L and a low expression level of E-cadherin compared to normal breast epithelial cells and non-TNBC cells. Here, a novel psammaplin A analog (PsA-3091) exhibited a potent inhibitory effect of DOT1L-mediated H3K79 methylation. Consistently, PsA-3091 also significantly inhibited the proliferation, migration, and invasion of TNBC cells along with the augmented expression of E-cadherin and the suppression of N-cadherin, ZEB1, and vimentin expression. In an orthotopic mouse model, PsA-3091 effectively inhibited lung metastasis and tumor growth by the regulation of DOT1L activity and EMT biomarkers. Together, we report here a new template of DOT1L inhibitor and suggest that targeting DOT1L-mediated H3K79 methylation by a novel PsA analog may be a promising strategy for the treatment of metastatic breast cancer patients.
Collapse
Affiliation(s)
- Woong Sub Byun
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Won Kyung Kim
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Hae Ju Han
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hwa-Jin Chung
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyungkuk Jang
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Han Sun Kim
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Sunghwa Kim
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Donghwa Kim
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun Seo Bae
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Sunghyouk Park
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeeyeon Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyeung-geun Park
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Sang Kook Lee
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
216
|
Zhao J, Jiang X, Yan L, Lin J, Guo H, Yu S, Ye B, Zhu J, Zhang W. Retinoic acid inducible gene-I slows down cellular senescence through negatively regulating the integrin β3/p38 MAPK pathway. Cell Cycle 2019; 18:3378-3392. [PMID: 31595820 PMCID: PMC6927694 DOI: 10.1080/15384101.2019.1677074] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 09/09/2019] [Accepted: 09/24/2019] [Indexed: 02/08/2023] Open
Abstract
Retinoic acid inducible gene-I (Rig-I) has been well documented as a cytosolic pattern recognition receptor that can sense viral RNA ligands to initiate the interferon-mediated antiviral immunity. However, little is known about the biological behaviors of Rig-I devoid of viral infection. Herein, we investigated the roles of Rig-I in the regulation of cellular senescence. In comparison to wild-type counterparts, Rig-I-/- mice displayed the accelerated loss of hair, less responsiveness to gentle physical stimuli and shorten survival time. Likewise, Rig-I deficiency rendered mouse embryonic fibroblasts (MEFs) more susceptible to the serial passages-associated replicative senescence. By performing a transcriptome analysis, we identified integrins at the intersections of biological pathways affected by Rig-I. Among these, integrin β3 was negatively regulated by Rig-I, and significantly upregulated with the occurrence of senescence. Gene silencing of Itgb3 (encoding integrin β3) retarded the progression of cellular senescence in both WT and Rig-I-/- MEFs. Notably, this effect was more prominent in Rig-I-/- MEFs. Furthermore, p38 MAPK was a key downstream molecule for integrin β3-mediated senescence, and overactivated in senescent Rig-I-/- MEFs. Taken together, Rig-I deficiency contributes to cellular senescence through amplifying integrin β3/p38 MAPK signaling. Our findings provide the evidence that Rig-I is a key regulator of cellular senescence, which will be helpful in better understanding its function without viral infection.Abbreviations: Rig-I: retinoic acid inducible gene-I; SASP: senescence-associated secretory phenotype; ECM: extracellular matrix; Itgb3: integrin beta 3; PRR: pattern recognition receptor; MEFs: mouse embryonic fibroblasts; Il-1β: interleukin-1 beta; Il-6: interleukin-6; Il-8: interleukin-8; Cxcl1: chemokine (C-X-C motif) ligand 1; Ccl2: chemokine (C-C motif) ligand 2; WT, wild type; BM: bone marrow; MAPK: mitogen-activated protein kinase; ERK: extracellular signal-regulated kinases; JNK: Jun N-terminal kinases; SA-β-gal: senescence-associated β-galactosidase; qPCR: quantitative reverse-transcription PCR; PBS: phosphate-buffered saline.
Collapse
Affiliation(s)
- Junmei Zhao
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology and Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- Department of Hematology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xinyi Jiang
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology and Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Li Yan
- Department of Hematology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Jian Lin
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology and Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- School of Life Sciences and Biotechnology, Shanghai Jiao-Tong University, Shanghai, China
| | - Hezhou Guo
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology and Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- School of Life Sciences and Biotechnology, Shanghai Jiao-Tong University, Shanghai, China
| | - Shanhe Yu
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology and Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Baixin Ye
- Department of Hematology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Jiang Zhu
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology and Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- School of Life Sciences and Biotechnology, Shanghai Jiao-Tong University, Shanghai, China
| | - Wu Zhang
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology and Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| |
Collapse
|
217
|
Wu X, Ding M, Lin J. Three-microRNA expression signature predicts survival in triple-negative breast cancer. Oncol Lett 2019; 19:301-308. [PMID: 31897142 PMCID: PMC6923981 DOI: 10.3892/ol.2019.11118] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a specific type of breast cancer with poor overall survival (OS) time. Previous studies revealed that microRNAs (miRNAs/miRs) serve important roles in the pathogenesis, progression and prognosis of TNBC. The present study analyzed the miRNA expression and clinical data of patients with TNBC downloaded from The Cancer Genome Atlas. A total of 194 differentially expressed miRNAs were identified between TNBC and matched normal tissues using the cut-off criteria of P<0.05 and |log2 fold change|>2. Of these miRNAs, 65 were downregulated and 129 were upregulated. Using Kaplan-Meier survival analysis, a total of 77 miRNAs that were closely associated with OS time were identified (P<0.05). The intersection of the 77 miRNAs and 194 differentially expressed miRNAs revealed six miRNAs. Log-rank tests based on survival curves were performed and two miRNAs were eliminated. The prognostic value of the remaining four miRNAs was evaluated with a Cox proportional hazards model using multiple logistic regression with forward stepwise selection of variables. Three miRNAs (miR-21-3p, miR-659-5p and miR-200b-5p) were subsequently identified as independent risk factors associated with OS time in the model. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses revealed that the target genes of these three miRNAs were mainly involved in ‘cell protein metabolism’, ‘RNA transcriptional regulation’, ‘cell migration’, ‘MAPK signaling pathway’, ‘ErbB signaling pathway’, ‘prolactin signaling pathway’ and ‘adherens junctions’. Taken together, the results obtained in the present study suggested that the three-miRNA signature may serve as a prognostic biomarker for patients with TNBC.
Collapse
Affiliation(s)
- Xinquan Wu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Mingji Ding
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Jianqin Lin
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| |
Collapse
|
218
|
He J, Ye W, Kou N, Chen K, Cui B, Zhang X, Hu S, Liu T, Kang L, Li X. MicroRNA-29b-3p suppresses oral squamous cell carcinoma cell migration and invasion via IL32/AKT signalling pathway. J Cell Mol Med 2019; 24:841-849. [PMID: 31680452 PMCID: PMC6933408 DOI: 10.1111/jcmm.14794] [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: 07/18/2019] [Revised: 09/04/2019] [Accepted: 10/09/2019] [Indexed: 12/17/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is aggressive accompanied with poor prognosis. We previously isolated the most invasive cells resembling the invasive tumour front by microfluidic technology and explored their differentially expressed microRNAs (miRNAs) in our previous work. Here, we verified the miR‐29b‐3p as a guarder that suppressed migration and invasion of OSCC cells and was down‐regulated in the most invasive cells. Besides that, the invasion suppression role of miR‐29b‐3p was achieved through the IL32/AKT pathway. Thus, miR‐29b‐3p and IL32 might serve as therapeutic targets for blocking the progression and improving the outcome of OSCC.
Collapse
Affiliation(s)
- Jianya He
- Department of Prosthodontics, College of Stomatology, Dalian Medical University, Dalian, China
| | - Wen Ye
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Ni Kou
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Dalian Medical University, Dalian, China
| | - Kang Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xiaohong Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Shuhai Hu
- Department of Prosthodontics, College of Stomatology, Dalian Medical University, Dalian, China
| | - Tingjiao Liu
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, Dalian, China
| | - Lan Kang
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaojie Li
- Department of Prosthodontics, College of Stomatology, Dalian Medical University, Dalian, China
| |
Collapse
|
219
|
Li J, Yang C, Yang J, Zou L. Down-regulation of CCL17 in cancer-associated fibroblasts inhibits cell migration and invasion of breast cancer through ERK1/2 pathway. Cancer Manag Res 2019; 11:7439-7453. [PMID: 31496803 PMCID: PMC6689663 DOI: 10.2147/cmar.s211651] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/17/2019] [Indexed: 02/05/2023] Open
Abstract
Objective Cancer-associated fibroblasts (CAFs) in the tumor microenvironment are involved in cancer development and progression, including breast cancer (BC). Up-regulation of CCL17 was observed in BC and predicted a decrease in overall survival, suggesting an important role of CCL17 in BC development. Nonetheless, little is known about the role of CCL17 in the interaction between CAFs and BC. Materials and methods Real-time quantitative PCR, Western blot, and enzyme-linked immunosorbent assay were performed to examine C-C motif chemokine ligand 17 (CCL17) and C-C motif chemokine receptor 4 (CCR4) levels in BC tissues and CAFs. Cell proliferation, migration, and invasion of CAFs co-cultured with or without BC cell lines were measured by Cell Counting Kit-8 and Transwell analysis. Expression of CCL17, CCR4, dual specificity phosphatase 6 (DUSP6), matrix metallopeptidase 13 (MMP13), extracellular signal-regulated kinase (ERK) 1/2, and phosphor-ERK1/2 (p-ERK1/2) in BC cell lines co-cultured with or without CAFs was measured by Western blotting. Results We found that BC tissues and CAFs demonstrated higher levels of CCL17 compared with adjacent-normal breast tissues and adjacent-normal fibroblasts (NFs), respectively. CCL17 expression is correlated with lymph nodes, TNM stage and tumor size of BC patients. CCL17 knockdown significantly inhibited CCL17 release, CCR4 expression, and the cell proliferation of CAFs, while CCL17 overexpression demonstrated an inverse effect in NFs. Co-culture with CAFs induced the increases in cell proliferation, migration, invasion, and the expression of CCL17, CCR4, MMP13, and p-ERK1/2 in MCF-7 and MDA-MB-231 cells were markedly reversed by CCL17 knockdown in CAFs. Meanwhile, co-culture with NFs induced the malignant phenotype of MCF-7 cells was markedly enhanced by CCL17 overexpression in NFs. Moreover, DUSP6, a negative regulator of ERK1/2, was dose-dependent decrease in response to recombinant CCL17 and inhibited cell migration, invasion, MMP13 expression, and ERK1/2 activation in MCF-7 cells. Conclusion The findings of this study suggest that CCL17 may function as a novel biomarker as well as potential therapeutic target against BC and CAF-secreted CCL17 promotes BC cell migration and invasion through the DUSP6-dependent ERK1/2 pathway.
Collapse
Affiliation(s)
- Junjie Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China.,Department of Breast Surgery, Sichuan Cancer Hospital, Chengdu 610041, People's Republic of China
| | - Chunli Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jingshi Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Liqun Zou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|
220
|
Hill BS, Sarnella A, D'Avino G, Zannetti A. Recruitment of stromal cells into tumour microenvironment promote the metastatic spread of breast cancer. Semin Cancer Biol 2019; 60:202-213. [PMID: 31377307 DOI: 10.1016/j.semcancer.2019.07.028] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023]
Abstract
Currently, metastasis remains the primary cause of death of patients with breast cancer despite the important advances in the treatment of this disease. In the complex tumour microenvironment network, several malignant and non-malignant cell types as well as components of extracellular matrix cooperate in promoting the metastatic spread of breast carcinoma. Many components of the stromal compartment are recruited from distant sites to the tumour including mesenchymal stem cells, endothelial cells, macrophages and other immune cells whereas other cells such as fibroblasts are already present in both primary and secondary lesions. When these cells come into contact with cancer cells they are "educated" and acquire a pro-tumoural phenotype, which support all the steps of the metastatic cascade. In this Review, we highlight the role played by each stromal component in guiding cancer cells in their venture towards colonizing metastatic sites.
Collapse
|
221
|
Chen Z, Yan X, Li K, Ling Y, Kang H. Stromal fibroblast-derived MFAP5 promotes the invasion and migration of breast cancer cells via Notch1/slug signaling. Clin Transl Oncol 2019; 22:522-531. [PMID: 31190277 DOI: 10.1007/s12094-019-02156-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The tumor microenvironment (TME) regulates tumor progression, and cancer-associated fibroblasts (CAFs) are the primary stromal components of the TME, with the potential to drive tumor metastasis via the secretion of paracrine factors, but the specific mechanisms driving this process have not been defined. METHODS Proteins secreted from CAFs and normal fibroblasts (NFs) were analyzed via proteomic analysis (fold change > 2, p < 0.05) to identify tumor-promoting proteins secreted by CAFs. RESULTS Proteomic analysis revealed that microfibrillar-associated protein 5 (MFAP5) is preferentially expressed and secreted by CAFs relative to NFs, which was confirmed by Western blotting and RT-qPCR. Transwell and wound healing assays confirmed that MFAP5 is secreted by CAFs, and drives the invasion and migration of MCF7 breast cancer cells. We further found that in MCF7 cells MFAP5 promoted epithelial-mesenchymal transition, activating Notch1 signaling and consequently upregulating NICD1 and slug. When Notch1 was knocked down in MCF7 cells, the ability of MFAP5 to promote invasion and migration decreased. CONCLUSION CAFs promote cancer cells invasion and migration via MFAP5 secretion and activation of the Notch1/slug signaling. These data highlight this pathway as a therapeutic target to disrupt tumor progression through the interference of CAF-tumor crosstalk.
Collapse
Affiliation(s)
- Z Chen
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - X Yan
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - K Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Y Ling
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - H Kang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
222
|
DiPersio CM, Van De Water L. Integrin Regulation of CAF Differentiation and Function. Cancers (Basel) 2019; 11:cancers11050715. [PMID: 31137641 PMCID: PMC6563118 DOI: 10.3390/cancers11050715] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/14/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023] Open
Abstract
Extensive remodeling of the extracellular matrix, together with paracrine communication between tumor cells and stromal cells, contribute to an “activated” tumor microenvironment that supports malignant growth and progression. These stromal cells include inflammatory cells, endothelial cells, and cancer-associated fibroblasts (CAFs). Integrins are expressed on all tumor and stromal cell types where they regulate both cell adhesion and bidirectional signal transduction across the cell membrane. In this capacity, integrins control pro-tumorigenic cell autonomous functions such as growth and survival, as well as paracrine crosstalk between tumor cells and stromal cells. The myofibroblast-like properties of cancer-associated fibroblasts (CAFs), such as robust contractility and extracellular matrix (ECM) deposition, allow them to generate both chemical and mechanical signals that support invasive tumor growth. In this review, we discuss the roles of integrins in regulating the ability of CAFs to generate and respond to extracellular cues in the tumor microenvironment. Since functions of specific integrins in CAFs are only beginning to emerge, we take advantage of a more extensive literature on how integrins regulate wound myofibroblast differentiation and function, as some of these integrin functions are likely to extrapolate to CAFs within the tumor microenvironment. In addition, we discuss the roles that integrins play in controlling paracrine signals that emanate from epithelial/tumor cells to stimulate fibroblasts/CAFs.
Collapse
|
223
|
Jang I, Beningo KA. Integrins, CAFs and Mechanical Forces in the Progression of Cancer. Cancers (Basel) 2019; 11:cancers11050721. [PMID: 31137693 PMCID: PMC6562616 DOI: 10.3390/cancers11050721] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 01/08/2023] Open
Abstract
Cells respond to both chemical and mechanical cues present within their microenvironment. Various mechanical signals are detected by and transmitted to the cells through mechanoreceptors. These receptors often contact with the extracellular matrix (ECM), where the external signals are converted into a physiological response. Integrins are well-defined mechanoreceptors that physically connect the actomyosin cytoskeleton to the surrounding matrix and transduce signals. Families of α and β subunits can form a variety of heterodimers that have been implicated in cancer progression and differ among types of cancer. These heterodimers serve as the nexus of communication between the cells and the tumor microenvironment (TME). The TME is dynamic and composed of stromal cells, ECM and associated soluble factors. The most abundant stromal cells within the TME are cancer-associated fibroblasts (CAFs). Accumulating studies implicate CAFs in cancer development and metastasis through their remodeling of the ECM and release of large amounts of ECM proteins and soluble factors. Considering that the communication between cancer cells and CAFs, in large part, takes place through the ECM, the involvement of integrins in the crosstalk is significant. This review discusses the role of integrins, as the primary cell-ECM mechanoreceptors, in cancer progression, highlighting integrin-mediated mechanical communication between cancer cells and CAFs.
Collapse
Affiliation(s)
- Imjoo Jang
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA.
| | - Karen A Beningo
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA.
| |
Collapse
|
224
|
Medeiros B, Allan AL. Molecular Mechanisms of Breast Cancer Metastasis to the Lung: Clinical and Experimental Perspectives. Int J Mol Sci 2019; 20:E2272. [PMID: 31071959 PMCID: PMC6540248 DOI: 10.3390/ijms20092272] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/01/2019] [Accepted: 05/06/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer in women worldwide, and >90% of breast cancer-related deaths are associated with metastasis. Breast cancer spreads preferentially to the lung, brain, bone and liver; termed organ tropism. Current treatment methods for metastatic breast cancer have been ineffective, compounded by the lack of early prognostic/predictive methods to determine which organs are most susceptible to developing metastases. A better understanding of the mechanisms that drive breast cancer metastasis is crucial for identifying novel biomarkers and therapeutic targets. Lung metastasis is of particular concern as it is associated with significant patient morbidity and a mortality rate of 60-70%. This review highlights the current understanding of breast cancer metastasis to the lung, including discussion of potential new treatment approaches for development.
Collapse
Affiliation(s)
- Braeden Medeiros
- London Regional Cancer Program, London Health Sciences Centre, Department of Anatomy & Cell Biology, Western University, London, ON N6A 5W9, Canada.
| | - Alison L Allan
- London Regional Cancer Program, London Health Sciences Centre, Departments of Anatomy & Cell Biology and Oncology, Western University, London, ON N6A 5W9, Canada.
| |
Collapse
|
225
|
Wu M, Wu W, Ding J, Yang J. LINC01433/miR-2116-3p/MYC Feedback Loop Promotes Cell Proliferation, Migration, and the Epithelial-Mesenchymal Transition in Breast Cancer. Cancer Biother Radiopharm 2019; 34:388-397. [PMID: 30939038 DOI: 10.1089/cbr.2019.2772] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background: It is reported that long noncoding RNAs play an important role in human cancers, including breast cancer (BC). However, the effect of long intergenic non-protein coding RNA 1433 (LINC01433) on BC development remains elusive. Materials and Methods: The expression level of LINC01433 in BC cells and a normal breast epithelial cell (MCF-10A) was determined by quantitative real-time polymerase chain reaction (qRT-PCR). A series of functional assays was applied to measure the bio-function of LINC01433 in BC. Bioinformatics analysis and mechanistic assays were utilized to disclose the underlying mechanism involved in the LINC01433-mediated BC cellular process. Results: qRT-PCR revealed that LINC01433 was highly expressed in BC cells. In function, LINC01433 depletion suppressed BC cell proliferation, migration, and epithelial-mesenchymal transition, but induced cell apoptosis. Mechanically, chromatin immunoprecipitation and luciferase reporter assays suggested that LINC01433 was activated by its upstream transcription factor MYC proto-oncogene (MYC). The interaction between LINC01433 and miR-2116-3p was verified in BC. Additionally, MYC was validated as a target gene of miR-2116-3p. Rescue assays demonstrated that LINC01433 promoted BC cellular process via regulating miR-2116-3p/MYC axis. Conclusion: Our findings revealed a novel positive feedback loop (LINC01433/miR-2116-3p/MYC) in BC progression and discovered the novel functional genes in this BC cellular process.
Collapse
Affiliation(s)
- Minhua Wu
- Department of Thyroid and Breast, Ningbo Medical Center Lihuili Eastern Hospital/Taipei Medical University Ningbo Medical Center, Ningbo, China
| | - Weizhu Wu
- Department of Thyroid and Breast, Ningbo Medical Center Lihuili Eastern Hospital/Taipei Medical University Ningbo Medical Center, Ningbo, China
| | - Jinhua Ding
- Department of Thyroid and Breast, Ningbo Medical Center Lihuili Eastern Hospital/Taipei Medical University Ningbo Medical Center, Ningbo, China
| | - Jiahui Yang
- Department of Thyroid and Breast, Ningbo Medical Center Lihuili Eastern Hospital/Taipei Medical University Ningbo Medical Center, Ningbo, China
| |
Collapse
|
226
|
The Role of Platelets in the Tumor-Microenvironment and the Drug Resistance of Cancer Cells. Cancers (Basel) 2019; 11:cancers11020240. [PMID: 30791448 PMCID: PMC6406993 DOI: 10.3390/cancers11020240] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 01/29/2019] [Accepted: 02/15/2019] [Indexed: 02/06/2023] Open
Abstract
Besides the critical functions in hemostasis, thrombosis and the wounding process, platelets have been increasingly identified as active players in various processes in tumorigenesis, including angiogenesis and metastasis. Once activated, platelets can release bioactive contents such as lipids, microRNAs, and growth factors into the bloodstream, subsequently enhancing the platelet⁻cancer interaction and stimulating cancer metastasis and angiogenesis. The mechanisms of treatment failure of chemotherapeutic drugs have been investigated to be associated with platelets. Therefore, understanding how platelets contribute to the tumor microenvironment may potentially identify strategies to suppress cancer angiogenesis, metastasis, and drug resistance. Herein, we present a review of recent investigations on the role of platelets in the tumor-microenvironment including angiogenesis, and metastasis, as well as targeting platelets for cancer treatment, especially in drug resistance.
Collapse
|
227
|
Therapeutic Potential of a Novel α vβ₃ Antagonist to Hamper the Aggressiveness of Mesenchymal Triple Negative Breast Cancer Sub-Type. Cancers (Basel) 2019; 11:cancers11020139. [PMID: 30682838 PMCID: PMC6406933 DOI: 10.3390/cancers11020139] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/17/2019] [Accepted: 01/20/2019] [Indexed: 12/20/2022] Open
Abstract
The mesenchymal sub-type of triple negative breast cancer (MES-TNBC) has a highly aggressive behavior and worse prognosis, due to its invasive and stem-like features, that correlate with metastatic dissemination and resistance to therapies. Furthermore, MES-TNBC is characterized by the expression of molecular markers related to the epithelial-to-mesenchymal transition (EMT) program and cancer stem cells (CSCs). The altered expression of αvβ3 integrin has been well established as a driver of cancer progression, stemness, and metastasis. Here, we showed that the high levels of αvβ3 are associated with MES-TNBC and therefore exploited the possibility to target this integrin to reduce the aggressiveness of this carcinoma. To this aim, MES-TNBC cells were treated with a novel peptide, named ψRGDechi, that we recently developed and characterized for its ability to selectively bind and inhibit αvβ3 integrin. Notably, ψRGDechi was able to hamper adhesion, migration, and invasion of MES-TNBC cells, as well as the capability of these cells to form vascular-like structures and mammospheres. In addition, this peptide reversed EMT program inhibits mesenchymal markers. These findings show that targeting αvβ3 integrin by ψRGDechi, it is possible to inhibit some of the malignant properties of MES-TNBC phenotype.
Collapse
|