1
|
Samaržija I. The Potential of Extracellular Matrix- and Integrin Adhesion Complex-Related Molecules for Prostate Cancer Biomarker Discovery. Biomedicines 2023; 12:79. [PMID: 38255186 PMCID: PMC10813710 DOI: 10.3390/biomedicines12010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/16/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Prostate cancer is among the top five cancer types according to incidence and mortality. One of the main obstacles in prostate cancer management is the inability to foresee its course, which ranges from slow growth throughout years that requires minimum or no intervention to highly aggressive disease that spreads quickly and resists treatment. Therefore, it is not surprising that numerous studies have attempted to find biomarkers of prostate cancer occurrence, risk stratification, therapy response, and patient outcome. However, only a few prostate cancer biomarkers are used in clinics, which shows how difficult it is to find a novel biomarker. Cell adhesion to the extracellular matrix (ECM) through integrins is among the essential processes that govern its fate. Upon activation and ligation, integrins form multi-protein intracellular structures called integrin adhesion complexes (IACs). In this review article, the focus is put on the biomarker potential of the ECM- and IAC-related molecules stemming from both body fluids and prostate cancer tissue. The processes that they are involved in, such as tumor stiffening, bone turnover, and communication via exosomes, and their biomarker potential are also reviewed.
Collapse
Affiliation(s)
- Ivana Samaržija
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
2
|
Macke AJ, Pachikov AN, Divita TE, Morris ME, LaGrange CA, Holzapfel MS, Kubyshkin AV, Zyablitskaya EY, Makalish TP, Eremenko SN, Qiu H, Riethoven JJM, Hemstreet GP, Petrosyan AA. Targeting the ATF6-Mediated ER Stress Response and Autophagy Blocks Integrin-Driven Prostate Cancer Progression. Mol Cancer Res 2023; 21:958-974. [PMID: 37314749 PMCID: PMC10527559 DOI: 10.1158/1541-7786.mcr-23-0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/27/2023] [Accepted: 06/09/2023] [Indexed: 06/15/2023]
Abstract
Prostate cancer progression to the lethal metastatic castration-resistant phenotype (mCRPC) is driven by αv integrins and is associated with Golgi disorganization and activation of the ATF6 branch of unfolded protein response (UPR). Overexpression of integrins requires N-acetylglucosaminyltransferase-V (MGAT5)-mediated glycosylation and subsequent cluster formation with Galectin-3 (Gal-3). However, the mechanism underlying this altered glycosylation is missing. For the first time, using HALO analysis of IHC, we found a strong association of integrin αv and Gal-3 at the plasma membrane (PM) in primary prostate cancer and mCRPC samples. We discovered that MGAT5 activation is caused by Golgi fragmentation and mislocalization of its competitor, N-acetylglucosaminyltransferase-III, MGAT3, from Golgi to the endoplasmic reticulum (ER). This was validated in an ethanol-induced model of ER stress, where alcohol treatment in androgen-refractory PC-3 and DU145 cells or alcohol consumption in patient with prostate cancer samples aggravates Golgi scattering, activates MGAT5, and enhances integrin expression at PM. This explains known link between alcohol consumption and prostate cancer mortality. ATF6 depletion significantly blocks UPR and reduces the number of Golgi fragments in both PC-3 and DU145 cells. Inhibition of autophagy by hydroxychloroquine (HCQ) restores compact Golgi, rescues MGAT3 intra-Golgi localization, blocks glycan modification via MGAT5, and abrogates delivery of Gal-3 to the cell surface. Importantly, the loss of Gal-3 leads to reduced integrins at PM and their accelerated internalization. ATF6 depletion and HCQ treatment synergistically decrease integrin αv and Gal-3 expression and temper orthotopic tumor growth and metastasis. IMPLICATIONS Combined ablation of ATF6 and autophagy can serve as new mCRPC therapeutic.
Collapse
Affiliation(s)
- Amanda J. Macke
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA, 68198
- The Fred and Pamela Buffett Cancer Center, Omaha, NE, USA, 68198
| | - Artem N. Pachikov
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA, 68198
- The Fred and Pamela Buffett Cancer Center, Omaha, NE, USA, 68198
| | - Taylor E. Divita
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA, 68198
- The Fred and Pamela Buffett Cancer Center, Omaha, NE, USA, 68198
| | - Mary E. Morris
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA, 68198
| | - Chad A. LaGrange
- Division of Urologic Surgery, Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA, 68198
| | - Melissa S. Holzapfel
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA, 68198
| | - Anatoly V. Kubyshkin
- Department of Pathological Physiology, Medical Academy named after S. I. Georgievsky, V. I. Vernadsky Crimean Federal University, Simferopol, Russia, 295051
| | - Evgeniya Y. Zyablitskaya
- Laboratory of Molecular Biology, Medical Academy named after S. I. Georgievsky, V. I. Vernadsky Crimean Federal University, Simferopol, Russia, 295051
| | - Tatiana P. Makalish
- Laboratory of Molecular Biology, Medical Academy named after S. I. Georgievsky, V. I. Vernadsky Crimean Federal University, Simferopol, Russia, 295051
| | - Sergey N. Eremenko
- Saint Luc’s Clinique, V. I. Vernadsky Crimean Federal University, Simferopol, Russia, 295051
| | - Haowen Qiu
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE, USA, 68588
| | - Jean-Jack M. Riethoven
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE, USA, 68588
- Department of Statistics, University of Nebraska-Lincoln, Lincoln, NE, USA, 68588
| | - George P. Hemstreet
- Division of Urologic Surgery, Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA, 68198
- Omaha Western Iowa Health Care System Urology, VA Service, Department of Research Service, Omaha, NE, USA, 68105
| | - and Armen Petrosyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA, 68198
- The Fred and Pamela Buffett Cancer Center, Omaha, NE, USA, 68198
| |
Collapse
|
3
|
Tsai CC, Yang YCSH, Chen YF, Huang LY, Yang YN, Lee SY, Wang WL, Lee HL, Whang-Peng J, Lin HY, Wang K. Integrins and Actions of Androgen in Breast Cancer. Cells 2023; 12:2126. [PMID: 37681860 PMCID: PMC10486718 DOI: 10.3390/cells12172126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 09/09/2023] Open
Abstract
Androgen has been shown to regulate male physiological activities and cancer proliferation. It is used to antagonize estrogen-induced proliferative effects in breast cancer cells. However, evidence indicates that androgen can stimulate cancer cell growth in estrogen receptor (ER)-positive and ER-negative breast cancer cells via different types of receptors and different mechanisms. Androgen-induced cancer growth and metastasis link with different types of integrins. Integrin αvβ3 is predominantly expressed and activated in cancer cells and rapidly dividing endothelial cells. Programmed death-ligand 1 (PD-L1) also plays a vital role in cancer growth. The part of integrins in action with androgen in cancer cells is not fully mechanically understood. To clarify the interactions between androgen and integrin αvβ3, we carried out molecular modeling to explain the potential interactions of androgen with integrin αvβ3. The androgen-regulated mechanisms on PD-L1 and its effects were also addressed.
Collapse
Affiliation(s)
- Chung-Che Tsai
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (C.-C.T.); (Y.-F.C.)
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Chen S. H. Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 11031, Taiwan;
| | - Yi-Fong Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (C.-C.T.); (Y.-F.C.)
| | - Lin-Yi Huang
- Department of Pediatrics, E-DA Hospital, I-Shou University, Kaohsiung 82445, Taiwan; (L.-Y.H.); (Y.-N.Y.)
| | - Yung-Ning Yang
- Department of Pediatrics, E-DA Hospital, I-Shou University, Kaohsiung 82445, Taiwan; (L.-Y.H.); (Y.-N.Y.)
- School of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Sheng-Yang Lee
- Dentistry, Wan-Fang Medical Center, Taipei Medical University, Taipei 11031, Taiwan;
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Wen-Long Wang
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242, Taiwan;
| | - Hsin-Lun Lee
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110, Taiwan
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan
| | | | - Hung-Yun Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (C.-C.T.); (Y.-F.C.)
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan;
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| | - Kuan Wang
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
4
|
Prigol AN, Rode MP, da Luz Efe F, Saleh NA, Creczynski-Pasa TB. The Bone Microenvironment Soil in Prostate Cancer Metastasis: An miRNA Approach. Cancers (Basel) 2023; 15:4027. [PMID: 37627055 PMCID: PMC10452124 DOI: 10.3390/cancers15164027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/28/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Bone metastatic prostate cancer (PCa) is associated with a high risk of mortality. Changes in the expression pattern of miRNAs seem to be related to early aspects of prostate cancer, as well as its establishment and proliferation, including the necessary steps for metastasis. Here we compiled, for the first time, the important roles of miRNAs in the development, diagnosis, and treatment of bone metastasis, focusing on recent in vivo and in vitro studies. PCa exosomes are proven to promote metastasis-related events, such as osteoblast and osteoclast differentiation and proliferation. Aberrant miRNA expression in PCa may induce abnormal bone remodeling and support tumor development. Furthermore, miRNAs are capable of binding to multiple mRNA targets, a dynamic property that can be harnessed for the development of treatment tools, such as antagomiRs and miRNA mimics, which have emerged as promising candidates in PCa treatment. Finally, miRNAs may serve as noninvasive biomarkers, as they can be detected in tissue and bodily fluids, are highly stable, and show differential expression between nonmetastatic PCa and bone metastatic samples. Taken together, the findings underscore the importance of miRNA expression profiles and miRNA-based tools as rational technologies to increase the quality of life and longevity of patients.
Collapse
Affiliation(s)
| | | | | | | | - Tânia Beatriz Creczynski-Pasa
- Department of Pharmaceutical Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, Santa Catarina State, Brazil; (A.N.P.); (M.P.R.); (F.d.L.E.); (N.A.S.)
| |
Collapse
|
5
|
Mao L, Wang L, Xu J, Zou J. The role of integrin family in bone metabolism and tumor bone metastasis. Cell Death Discov 2023; 9:119. [PMID: 37037822 PMCID: PMC10086008 DOI: 10.1038/s41420-023-01417-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023] Open
Abstract
Integrins have been the research focus of cell-extracellular matrix adhesion (ECM) and cytokine receptor signal transduction. They are involved in the regulation of bone metabolism of bone precursor cells, mesenchymal stem cells (MSCs), osteoblasts (OBs), osteoclasts (OCs), and osteocytes. Recent studies expanded and updated the role of integrin in bone metabolism, and a large number of novel cytokines were found to activate bone metabolism pathways through interaction with integrin receptors. Integrins act as transducers that mediate the regulation of bone-related cells by mechanical stress, fluid shear stress (FSS), microgravity, hypergravity, extracellular pressure, and a variety of physical factors. Integrins mediate bone metastasis of breast, prostate, and lung cancer by promoting cancer cell adhesion, migration, and survival. Integrin-mediated targeted therapy showed promising prospects in bone metabolic diseases. This review emphasizes the latest research results of integrins in bone metabolism and bone metastasis and provides a vision for treatment strategies.
Collapse
Affiliation(s)
- Liwei Mao
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China
| | - Lian Wang
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, WA, 6009, Perth, Australia
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China.
| |
Collapse
|
6
|
Nintedanib-αVβ6 Integrin Ligand Conjugates Reduce TGF β-Induced EMT in Human Non-Small Cell Lung Cancer. Int J Mol Sci 2023; 24:ijms24021475. [PMID: 36674990 PMCID: PMC9861180 DOI: 10.3390/ijms24021475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Growth factors and cytokines released in the lung cancer microenvironment promote an epithelial-to-mesenchymal transition (EMT) that sustains the progression of neoplastic diseases. TGFβ is one of the most powerful inducers of this transition, as it induces overexpression of the fibronectin receptor, αvβ6 integrin, in cancer cells which, in turn, is strongly associated with EMT. Thus, αvβ6 integrin receptors may be exploited as a target for the selective delivery of anti-tumor agents. We introduce three novel synthesized conjugates, in which a selective αvβ6 receptor ligand is linked to nintedanib, a potent kinase inhibitor used to treat advanced adenocarcinoma lung cancer in clinics. The αvβ6 integrin ligand directs nintedanib activity to the target cells of the tumor microenvironment, avoiding the onset of negative side effects in normal cells. We found that the three conjugates inhibit the adhesion of cancer cells to fibronectin in a concentration-dependent manner and that αvβ6-expressing cells internalized the conjugated compounds, thus permitting nintedanib to inhibit 2D and 3D cancer cell growth and suppress the clonogenic ability of the EMT phenotype as well as intervening in other aspects associated with the EMT transition. These results highlight αvβ6 receptors as privileged access points for dual-targeting molecular conjugates engaged in an efficient and precise strategy against non-small cell lung cancer.
Collapse
|
7
|
Chen C, Huang R, Zhou J, Guo L, Xiang S. Formation of pre-metastatic bone niche in prostate cancer and regulation of traditional chinese medicine. Front Pharmacol 2022; 13:897942. [PMID: 36059977 PMCID: PMC9428453 DOI: 10.3389/fphar.2022.897942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022] Open
Abstract
Prostate cancer with bone metastasis has a high cancer-specific mortality. Thus, it is essential to delineate the mechanism of bone metastasis. Pre-metastatic niche (PMN) is a concept in tumor metastasis, which is characterized by tumor-secreted factors, reprogramming of stromal cells, and immunosuppression by myeloid-derived suppressor cells (MDSC), which is induced by bone marrow-derived cells (BMDC) in the target organ. However, PMN does not explain the predilection of prostate cancer towards bone metastasis. In this review, we discuss the initiation of bone metastasis of prostate cancer from the perspective of PMN and tumor microenvironment in a step-wise manner. Furthermore, we present a new concept called pre-metastatic bone niche, featuring inherent BMDC, to interpret bone metastasis. Moreover, we illustrate the regulation of traditional Chinese medicine on PMN.
Collapse
|
8
|
Krishn SR, Garcia V, Naranjo NM, Quaglia F, Shields CD, Harris MA, Kossenkov AV, Liu Q, Corey E, Altieri DC, Languino LR. Small extracellular vesicle-mediated ITGB6 siRNA delivery downregulates the αVβ6 integrin and inhibits adhesion and migration of recipient prostate cancer cells. Cancer Biol Ther 2022; 23:173-185. [PMID: 35188070 PMCID: PMC8865252 DOI: 10.1080/15384047.2022.2030622] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The αVβ6 integrin, an epithelial-specific cell surface receptor absent in normal prostate and expressed during prostate cancer (PrCa) progression, is a therapeutic target in many cancers. Here, we report that transcript levels of ITGB6 (encoding the β6 integrin subunit) are significantly increased in metastatic castrate-resistant androgen receptor-negative prostate tumors compared to androgen receptor-positive prostate tumors. In addition, the αVβ6 integrin protein levels are significantly elevated in androgen receptor-negative PrCa patient derived xenografts (PDXs) compared to androgen receptor-positive PDXs. In vitro, the androgen receptor-negative PrCa cells express high levels of the αVβ6 integrin compared to androgen receptor-positive PrCa cells. Additionally, expression of androgen receptor (wild type or variant 7) in androgen receptor-negative PrCa cells downregulates the expression of the β6 but not αV subunit compared to control cells. We demonstrate an efficient strategy to therapeutically target the αVβ6 integrin during PrCa progression by using short interfering RNA (siRNA) loaded into PrCa cell-derived small extracellular vesicles (sEVs). We first demonstrate that fluorescently-labeled siRNAs can be efficiently loaded into PrCa cell-derived sEVs by electroporation. By confocal microscopy, we show efficient internalization of these siRNA-loaded sEVs into PrCa cells. We show that sEV-mediated delivery of ITGB6-targeting siRNAs into PC3 cells specifically downregulates expression of the β6 subunit. Furthermore, treatment with sEVs encapsulating ITGB6 siRNA significantly reduces cell adhesion and migration of PrCa cells on an αVβ6-specific substrate, LAP-TGFβ1. Our results demonstrate an approach for specific targeting of the αVβ6 integrin in PrCa cells using sEVs encapsulating ITGB6-specific siRNAs.
Collapse
Affiliation(s)
- Shiv Ram Krishn
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA USA
| | - Vaughn Garcia
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA USA
| | - Nicole M. Naranjo
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA USA
| | - Fabio Quaglia
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA USA
| | - Christopher D. Shields
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA USA
| | - Maisha A. Harris
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA USA
| | - Andrew V. Kossenkov
- Center for Systems and Computational Biology, the Wistar Institute, Philadelphia, PA USA
| | - Qin Liu
- Molecular and Cellular Oncogenesis Program, the Wistar Institute, Philadelphia, PA USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA USA
| | - Dario C. Altieri
- Prostate Cancer Discovery and Development Program, the Wistar Institute, Philadelphia, PA USA
- Immunology, Microenvironment and Metastasis Program, the Wistar Institute, Philadelphia, PA USA
| | - Lucia R. Languino
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA USA
| |
Collapse
|
9
|
Abstract
The transmission of information between tumor cells and other cell types in the tumor microenvironment plays an important role in tumor metastasis and is critically modulated by exosomes and other mediators. Tumor-derived exosomes can promote epithelial-mesenchymal transition, angiogenesis, immune escape, formation of the pre-metastatic microenvironment, and transmission of drug-resistant molecules, thereby promoting tumor growth, invasion, and metastasis. Integrins are important regulatory molecules on exosomes that can locate metastatic cells at the initial stage of metastasis and show good organotropism. This fact suggests that a clear understanding of the roles of exosomal integrins will be beneficial for future clinical applications. Follow-up studies on exosomes using continuously updated purification techniques and identification methods are extremely important. In addition to their potential as cancer biomarkers, exosomes also provide new research directions for precision medicine. Currently, exosomes have potential value in disease treatment and provide clinicians with more meaningful judgment standards.
Collapse
|
10
|
Quaglia F, Krishn SR, Wang Y, Goodrich DW, McCue P, Kossenkov AV, Mandigo AC, Knudsen KE, Weinreb PH, Corey E, Kelly WK, Languino LR. Differential expression of αVβ3 and αVβ6 integrins in prostate cancer progression. PLoS One 2021; 16:e0244985. [PMID: 33481853 PMCID: PMC7822502 DOI: 10.1371/journal.pone.0244985] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022] Open
Abstract
Neuroendocrine prostate cancer (NEPrCa) arises de novo or after accumulation of genomic alterations in pre-existing adenocarcinoma tumors in response to androgen deprivation therapies. We have provided evidence that small extracellular vesicles released by PrCa cells and containing the αVβ3 integrin promote neuroendocrine differentiation of PrCa in vivo and in vitro. Here, we examined αVβ3 integrin expression in three murine models carrying a deletion of PTEN (SKO), PTEN and RB1 (DKO), or PTEN, RB1 and TRP53 (TKO) genes in the prostatic epithelium; of these three models, the DKO and TKO tumors develop NEPrCa with a gene signature comparable to those of human NEPrCa. Immunostaining analysis of SKO, DKO and TKO tumors shows that αVβ3 integrin expression is increased in DKO and TKO primary tumors and metastatic lesions, but absent in SKO primary tumors. On the other hand, SKO tumors show higher levels of a different αV integrin, αVβ6, as compared to DKO and TKO tumors. These results are confirmed by RNA-sequencing analysis. Moreover, TRAMP mice, which carry NEPrCa and adenocarcinoma of the prostate, also have increased levels of αVβ3 in their NEPrCa primary tumors. In contrast, the αVβ6 integrin is only detectable in the adenocarcinoma areas. Finally, analysis of 42 LuCaP patient-derived xenografts and primary adenocarcinoma samples shows a positive correlation between αVβ3, but not αVβ6, and the neuronal marker synaptophysin; it also demonstrates that αVβ3 is absent in prostatic adenocarcinomas. In summary, we demonstrate that αVβ3 integrin is upregulated in NEPrCa primary and metastatic lesions; in contrast, the αVβ6 integrin is confined to adenocarcinoma of the prostate. Our findings suggest that the αVβ3 integrin, but not αVβ6, may promote a shift in lineage plasticity towards a NE phenotype and might serve as an informative biomarker for the early detection of NE differentiation in prostate cancer.
Collapse
Affiliation(s)
- Fabio Quaglia
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States of America
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Shiv Ram Krishn
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States of America
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Yanqing Wang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States of America
| | - David W. Goodrich
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States of America
| | - Peter McCue
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Andrew V. Kossenkov
- Center for Systems and Computational Biology, Wistar Institute, Philadelphia, PA, United States of America
| | - Amy C. Mandigo
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Karen E. Knudsen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States of America
| | | | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, United States of America
| | - William K. Kelly
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Lucia R. Languino
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States of America
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States of America
| |
Collapse
|
11
|
Urquiza M, Guevara V, Diaz-Sana E, Mora F. The Role of αvβ6 Integrin Binding Molecules in the Diagnosis and Treatment of Cancer. CURR ORG CHEM 2020. [DOI: 10.2174/1385272824999200528124936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Peptidic and non-peptidic αvβ6 integrin-binding molecules have been used in
the clinic for detection and treatment of tumors expressing αvβ6 integrin, because this protein
is expressed in malignant epithelial cells of the oral cavity, pancreas, breast, ovary,
colon and stomach carcinomas but it is not expressed in healthy adult tissue except during
wound healing and inflammation. This review focuses on the landscape of αvβ6 integrinbinding
molecules and their use in cancer treatment and detection, and discusses recent
designs for tumor detection, treatment, and immunotherapy. In the last ten years, several
reviews abamp;#945;vβ6 integrin-binding molecules and their role in cancer detection and treatment.
Firstly, this review describes the role of the αvβ6 integrin in normal tissues, how the expression
of this protein is correlated with cancer severity and its role in cancer development. Taking into account
the potential of αvβ6 integrin-binding molecules in detection and treatment of specific tumors, special
attention is given to several high-affinity αvβ6 integrin-binding peptides used for tumor imaging; particularly,
the αvβ6-binding peptide NAVPNLRGDLQVLAQKVART [A20FMDV2], derived from the foot and mouth
disease virus. This peptide labeled with either 18F, 111In or with 68Ga has been used for PET imaging of αvβ6
integrin-positive tumors. Moreover, αvβ6 integrin-binding peptides have been used for photoacoustic and fluorescence
imaging and could potentially be used in clinical application in cancer diagnosis and intraoperative
imaging of αvβ6-integrin positive tumors. Additionally, non-peptidic αvβ6-binding molecules have been designed
and used in the clinic for the detection and treatment of αvβ6-expressing tumors. Anti-αvβ6 integrin antibodies
are another useful tool for selective identification and treatment of αvβ6 (+) tumors. The utility of
these αvβ6 integrin-binding molecules as a tool for tumor detection and treatment is discussed, considering
specificity, sensitivity and serum stability. Another use of the αvβ6 integrin-binding peptides is to modify the
Ad5 cell tropism for inducing oncolytic activity of αvβ6-integrin positive tumor cells by expressing
A20FMDV2 peptide within the fiber knob protein (Ad5NULL-A20). The newly designed oncolytic
Ad5NULL-A20 virotherapy is promising for local and systemic targeting of αvβ6-overexpressing cancers. Finally,
new evidence has emerged, indicating that chimeric antigen receptor (CAR) containing the αvβ6 integrin-
binding peptide on top of CD28+CD3 endodomain displays a potent therapeutic activity in a diverse
repertoire of solid tumor models.
Collapse
Affiliation(s)
- Mauricio Urquiza
- Grupo de Investigacion en Hormonas (GIH), Department of Chemistry, National University of Columbia, Cra 30 # 45-03, Bogota, zip code 111321, Colombia
| | - Valentina Guevara
- Grupo de Investigacion en Hormonas (GIH), Department of Chemistry, National University of Columbia, Cra 30 # 45-03, Bogota, zip code 111321, Colombia
| | - Erika Diaz-Sana
- Grupo de Investigacion en Hormonas (GIH), Department of Chemistry, National University of Columbia, Cra 30 # 45-03, Bogota, zip code 111321, Colombia
| | - Felipe Mora
- Grupo de Investigacion en Hormonas (GIH), Department of Chemistry, National University of Columbia, Cra 30 # 45-03, Bogota, zip code 111321, Colombia
| |
Collapse
|
12
|
Li X, Jin L, Tan Y. Different roles of matrix metalloproteinase 2 in osteolysis of skeletal dysplasia and bone metastasis (Review). Mol Med Rep 2020; 23:70. [PMID: 33236155 PMCID: PMC7716421 DOI: 10.3892/mmr.2020.11708] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/02/2020] [Indexed: 01/18/2023] Open
Abstract
Matrix metalloproteinase 2 (MMP2) is a well-characterized protein that is indispensable for extracellular matrix remodeling and other pathological processes, such as tumor progression and skeletal dysplasia. Excessive activation of MMP2 promotes osteolytic metastasis and bone destruction in late-stage cancers, while its loss-of-function mutations result in the decreased bone mineralization and generalized osteolysis occurring progressively in skeletal developmental disorders, particularly in multicentric osteolysis, nodulosis and arthropathy (MONA). Either upregulation or downregulation of MMP2 activity can result in the same osteolytic effects. Thus, different functions of MMP2 have been recently identified that could explain this observation. While MMP2 can degrade bone matrix, facilitate osteoclastogenesis and amplify various signaling pathways that enhance osteolysis in bone metastasis, its role in maintaining the number of bone cells, supporting osteocytic canalicular network formation and suppressing leptin-mediated inhibition of bone formation has been implicated in osteolytic disorders caused by MMP2 deficiency. Furthermore, the proangiogenic activity of MMP2 is one of the potential mechanisms that are associated with both pathological situations. In the present article, the latest research on MMP2 in bone homeostasis is reviewed and the mechanisms underlying the role of this protein in skeletal metastasis and developmental osteolysis are discussed.
Collapse
Affiliation(s)
- Xiumao Li
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Libin Jin
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yanbin Tan
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
13
|
Targeting IL-3Rα on tumor-derived endothelial cells blunts metastatic spread of triple-negative breast cancer via extracellular vesicle reprogramming. Oncogenesis 2020; 9:90. [PMID: 33040091 PMCID: PMC7548009 DOI: 10.1038/s41389-020-00274-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/10/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
The lack of approved targeted therapies highlights the need for new treatments for triple-negative breast cancer (TNBC) patients. Interleukin-3 (IL-3) acts as an autocrine factor for tumor-endothelial cells (TEC), and exerts pro-angiogenic paracrine action via extracellular vesicles (EVs). IL-3Rα blockade on TEC changes TEC-EV (anti-IL-3R-EV) microRNA (miR) content and promotes the regression of established vessels. As TEC is the doorway for "drug" entry into tumors, we aimed to assess whether IL-3R blockade on TEC impacts tumor progression via its unique EV cargo. First, the expression of IL-3Rα was evaluated in 27 human TNBC samples. It was noticed that, besides TEC and inflammatory cells, tumor cells from 55.5% of the human TNBC samples expressed IL-3Rα. Using human TNBC cell lines for in vitro studies, we found that, unlike native TEC-EVs (nEVs), anti-IL-3R-EVs increase apoptosis and reduced cell viability and migration. In vivo, anti-IL-3R-EV treatment induced vessel regression in established tumors formed of MDA-MB-231 cells, decreased Vimentin, β-catenin, and TWIST1 expression, almost abolished liver and lung metastases from primary tumors, and reduced lung metastasis generated via the intravenous injection of MDA-MB-231 cells. nEVs depleted of miR-24-3p (antago-miR-24-3p-EVs) were effective as anti-IL-3R-EVs in downregulating TWIST1 and reducing metastatic lesions in vivo. Consistent with network analyses of miR-24-3p gene targeting, anti-IL-3R-EVs and antago-miR-24-3p-EVs upregulate SPRY2 in MDA-MB-231 cells. Finally, SPRY2 silencing prevented anti-IL-3R-EV and antago-miR-24-3p-EV-mediated apoptotic cues.Overall, these data provide the first evidence that IL-3Rα is highly expressed in TNBC cells, TEC, and inflammatory cells, and that IL-3Rα blockade on TEC impacts tumor progression.
Collapse
|
14
|
Krishn SR, Salem I, Quaglia F, Naranjo NM, Agarwal E, Liu Q, Sarker S, Kopenhaver J, McCue PA, Weinreb PH, Violette SM, Altieri DC, Languino LR. The αvβ6 integrin in cancer cell-derived small extracellular vesicles enhances angiogenesis. J Extracell Vesicles 2020; 9:1763594. [PMID: 32595914 PMCID: PMC7301698 DOI: 10.1080/20013078.2020.1763594] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 03/26/2020] [Accepted: 04/18/2020] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer (PrCa) cells crosstalk with the tumour microenvironment by releasing small extracellular vesicles (sEVs). sEVs, as well as large extracellular vesicles (LEVs), isolated via iodixanol density gradients from PrCa cell culture media, express the epithelial-specific αvβ6 integrin, which is known to be induced in cancer. In this study, we show sEV-mediated protein transfer of αvβ6 integrin to microvascular endothelial cells (human microvascular endothelial cells 1 - HMEC1) and demonstrate that de novo αvβ6 integrin expression is not caused by increased mRNA levels. Incubation of HMEC1 with sEVs isolated from PrCa PC3 cells that express the αvβ6 integrin results in a highly significant increase in the number of nodes, junctions and tubules. In contrast, incubation of HMEC1 with sEVs isolated from β6 negative PC3 cells, generated by shRNA against β6, results in a reduction in the number of nodes, junctions and tubules, a decrease in survivin levels and an increase in a negative regulator of angiogenesis, pSTAT1. Furthermore, treatment of HMEC1 with sEVs generated by CRISPR/Cas9-mediated down-regulation of β6, causes up-regulation of pSTAT1. Overall, our findings suggest that αvβ6 integrin in cancer sEVs regulates angiogenesis during PrCa progression.
Collapse
Affiliation(s)
- Shiv Ram Krishn
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, USA.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Israa Salem
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, USA.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Fabio Quaglia
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, USA.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Nicole M Naranjo
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, USA.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Ekta Agarwal
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, USA.,Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, USA
| | - Qin Liu
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, USA
| | - Srawasti Sarker
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, USA.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Jessica Kopenhaver
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Peter A McCue
- Department of Pathology, Thomas Jefferson University, Philadelphia, USA
| | | | | | - Dario C Altieri
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, USA.,Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, USA
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, USA.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| |
Collapse
|
15
|
Sun Q, Dong X, Shang Y, Sun F, Niu J, Li F. Integrin αvβ6 predicts poor prognosis and promotes resistance to cisplatin in hilar cholangiocarcinoma. Pathol Res Pract 2020; 216:153022. [PMID: 32534716 DOI: 10.1016/j.prp.2020.153022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/23/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Integrin αvβ6 is associated with an extremely aggressive cancer phenotype. However, little is known about the clinicopathological significance and prognostic value of integrin αvβ6 in human hilar cholangiocarcinoma. METHODS In the present study, bioinformatics analysis demonstrated a significant increase of integrin β6 gene expression in cholangiocarcinoma tissues compared to non-tumorous tissues, which was further validated in clinical samples through RT-qPCR and western blotting analyses. Integrin αvβ6 was observed to be expressed in 48.6% of tumors, and its expression was related to a poor tumor differentiation (p = 0.002), lymph node metastasis (p<0.001) and advanced TNM stage (p=0.001). Furthermore, patients who were αvβ6-positive showed a significantly shorter overall survival period than those who were αvβ6-negative (p=0.004). Multivariate analysis confirmed that integrin αvβ6 was an independent prognostic factor (p=0.002). In addition, loss- and gain-of-function assays showed integrin αvβ6 not only played an important role in colony formation, but also protected cholangiocarcinoma cells from cisplatin-induced growth inhibition and apoptosis. ERK/MAPK signaling pathway was involved in integrin αvβ6-mediated resistance of cholangiocarcinoma cells to cisplatin. CONCLUSIONS Taken together, the present findings revealed that integrin αvβ6 could serve as a potential prognostic predictor and contribute to cisplatin resistance, which might prove to be a promising target candidate for the clinical intervention of human hilar cholangiocarcinoma.
Collapse
Affiliation(s)
- Qi Sun
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiwen Dong
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yukui Shang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Fengkai Sun
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Jun Niu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Fanni Li
- Department of Talent Highland, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
16
|
Frisbie CP, Lushnikov AY, Krasnoslobodtsev AV, Riethoven JJM, Clarke JL, Stepchenkova EI, Petrosyan A. Post-ER Stress Biogenesis of Golgi Is Governed by Giantin. Cells 2019; 8:E1631. [PMID: 31847122 PMCID: PMC6953117 DOI: 10.3390/cells8121631] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The Golgi apparatus undergoes disorganization in response to stress, but it is able to restore compact and perinuclear structure under recovery. This self-organization mechanism is significant for cellular homeostasis, but remains mostly elusive, as does the role of giantin, the largest Golgi matrix dimeric protein. METHODS In HeLa and different prostate cancer cells, we used the model of cellular stress induced by Brefeldin A (BFA). The conformational structure of giantin was assessed by proximity ligation assay and atomic force microscopy. The post-BFA distribution of Golgi resident enzymes was examined by 3D SIM high-resolution microscopy. RESULTS We detected that giantin is rather flexible than an extended coiled-coil dimer and BFA-induced Golgi disassembly was associated with giantin monomerization. A fusion of the nascent Golgi membranes after BFA washout is forced by giantin re-dimerization via disulfide bond in its luminal domain and assisted by Rab6a GTPase. GM130-GRASP65-dependent enzymes are able to reach the nascent Golgi membranes, while giantin-sensitive enzymes appeared at the Golgi after its complete recovery via direct interaction of their cytoplasmic tail with N-terminus of giantin. CONCLUSION Post-stress recovery of Golgi is conducted by giantin dimer and Golgi proteins refill membranes according to their docking affiliation rather than their intra-Golgi location.
Collapse
Affiliation(s)
- Cole P. Frisbie
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA;
| | - Alexander Y. Lushnikov
- Nanoimaging Core Facility, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA; (A.Y.L.); (A.V.K.)
| | - Alexey V. Krasnoslobodtsev
- Nanoimaging Core Facility, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA; (A.Y.L.); (A.V.K.)
- Department of Physics, University of Nebraska-Omaha, Omaha, NE 68182-0266, USA
| | - Jean-Jack M. Riethoven
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68588-0665, USA;
- The Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE 68588-0304, USA;
| | - Jennifer L. Clarke
- The Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE 68588-0304, USA;
- Department of Statistics, University of Nebraska-Lincoln, Lincoln, NE 68583-0963, USA
| | - Elena I. Stepchenkova
- Vavilov Institute of General Genetics, Saint-Petersburg Branch, Russian Academy of Sciences, Saint-Petersburg 199034, Russia;
- Department of Genetics, Saint-Petersburg State University, Saint-Petersburg 199034, Russia
| | - Armen Petrosyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA;
- The Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE 68588-0304, USA;
- The Fred and Pamela Buffett Cancer Center, Omaha, NE 68198-5870, USA
| |
Collapse
|
17
|
Nath D, Li X, Mondragon C, Post D, Chen M, White JR, Hryniewicz-Jankowska A, Caza T, Kuznetsov VA, Hehnly H, Jamaspishvili T, Berman DM, Zhang F, Kung SHY, Fazli L, Gleave ME, Bratslavsky G, Pandolfi PP, Kotula L. Abi1 loss drives prostate tumorigenesis through activation of EMT and non-canonical WNT signaling. Cell Commun Signal 2019; 17:120. [PMID: 31530281 PMCID: PMC6749699 DOI: 10.1186/s12964-019-0410-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/01/2019] [Indexed: 12/29/2022] Open
Abstract
Background Prostate cancer development involves various mechanisms, which are poorly understood but pointing to epithelial mesenchymal transition (EMT) as the key mechanism in progression to metastatic disease. ABI1, a member of WAVE complex and actin cytoskeleton regulator and adaptor protein, acts as tumor suppressor in prostate cancer but the role of ABI1 in EMT is not clear. Methods To investigate the molecular mechanism by which loss of ABI1 contributes to tumor progression, we disrupted the ABI1 gene in the benign prostate epithelial RWPE-1 cell line and determined its phenotype. Levels of ABI1 expression in prostate organoid tumor cell lines was evaluated by Western blotting and RNA sequencing. ABI1 expression and its association with prostate tumor grade was evaluated in a TMA cohort of 505 patients and metastatic cell lines. Results Low ABI1 expression is associated with biochemical recurrence, metastasis and death (p = 0.038). Moreover, ABI1 expression was significantly decreased in Gleason pattern 5 vs. pattern 4 (p = 0.0025) and 3 (p = 0.0012), indicating an association between low ABI1 expression and highly invasive prostate tumors. Disruption of ABI1 gene in RWPE-1 cell line resulted in gain of an invasive phenotype, which was characterized by a loss of cell-cell adhesion markers and increased migratory ability of RWPE-1 spheroids. Through RNA sequencing and protein expression analysis, we discovered that ABI1 loss leads to activation of non-canonical WNT signaling and EMT pathways, which are rescued by re-expression of ABI1. Furthermore, an increase in STAT3 phosphorylation upon ABI1 inactivation and the evidence of a high-affinity interaction between the FYN SH2 domain and ABI1 pY421 support a model in which ABI1 acts as a gatekeeper of non-canonical WNT-EMT pathway activation downstream of the FZD2 receptor. Conclusions ABI1 controls prostate tumor progression and epithelial plasticity through regulation of EMT-WNT pathway. Here we discovered that ABI1 inhibits EMT through suppressing FYN-STAT3 activation downstream from non-canonical WNT signaling thus providing a novel mechanism of prostate tumor suppression. Electronic supplementary material The online version of this article (10.1186/s12964-019-0410-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Disharee Nath
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Xiang Li
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Claudia Mondragon
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA
| | - Dawn Post
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA
| | - Ming Chen
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.,Present address: Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA.,Duke Cancer Institute, Duke University, Durham, NC, 27710, USA
| | - Julie R White
- Laboratory of Comparative Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Anita Hryniewicz-Jankowska
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA.,Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Tiffany Caza
- Department of Pathology and Medicine, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Vladimir A Kuznetsov
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA.,Bioinformatics Institute, A-STAR, Singapore, 138671, Singapore
| | - Heidi Hehnly
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Tamara Jamaspishvili
- Department of Pathology and Molecular Medicine and Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, 10 Stuart St, Kingston, ON, K7L 3N6, Canada
| | - David M Berman
- Department of Pathology and Molecular Medicine and Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, 10 Stuart St, Kingston, ON, K7L 3N6, Canada
| | - Fan Zhang
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H 3Z6, Canada
| | - Sonia H Y Kung
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H 3Z6, Canada
| | - Ladan Fazli
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H 3Z6, Canada
| | - Martin E Gleave
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H 3Z6, Canada
| | - Gennady Bratslavsky
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Leszek Kotula
- Department of Urology, Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York, 13210, USA. .,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
18
|
Integrin-Mediated TGFβ Activation Modulates the Tumour Microenvironment. Cancers (Basel) 2019; 11:cancers11091221. [PMID: 31438626 PMCID: PMC6769837 DOI: 10.3390/cancers11091221] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/12/2019] [Accepted: 08/15/2019] [Indexed: 12/26/2022] Open
Abstract
TGFβ (transforming growth factor-beta) is a pleotropic cytokine with contrasting effects in cancer. In normal tissue and early tumours, TGFβ acts as a tumour suppressor, limiting proliferation and inducing apoptosis. However, these effects are eventually abrogated by the loss or inactivation of downstream signalling within the TGFβ pathway, and in established tumours, TGFβ then acts as a tumour promotor through multiple mechanisms including inducing epithelial-to-mesenchymal transition (EMT), promoting formation of cancer-associated fibroblasts (CAFs) and increasing angiogenesis. TGFβ is secrereted as a large latent complex and is embedded in the extracellular matrix or held on the surface of cells and must be activated before mediating its multiple functions. Thus, whilst TGFβ is abundant in the tumour microenvironment (TME), its functionality is regulated by local activation. The αv-integrins are major activators of latent-TGFβ. The potential benefits of manipulating the immune TME have been highlighted by the clinical success of immune-checkpoint inhibitors in a number of solid tumour types. TGFβ is a potent suppressor of T-cell-mediated immune surveillance and a key cause of resistance to checkpoint inhibitors. Therefore, as certain integrins locally activate TGFβ, they are likely to have a role in the immunosuppressive TME, although this remains to be confirmed. In this review, we discussed the role of TGFβ in cancer, the role of integrins in activating TGFβ in the TME, and the potential benefits of targeting integrins to augment immunotherapies.
Collapse
|
19
|
Ciardiello C, Leone A, Lanuti P, Roca MS, Moccia T, Minciacchi VR, Minopoli M, Gigantino V, De Cecio R, Rippa M, Petti L, Capone F, Vitagliano C, Milone MR, Pucci B, Lombardi R, Iannelli F, Di Gennaro E, Bruzzese F, Marchisio M, Carriero MV, Di Vizio D, Budillon A. Large oncosomes overexpressing integrin alpha-V promote prostate cancer adhesion and invasion via AKT activation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:317. [PMID: 31319863 PMCID: PMC6639931 DOI: 10.1186/s13046-019-1317-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/09/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Molecular markers for prostate cancer (PCa) are required to improve the early definition of patient outcomes. Atypically large extracellular vesicles (EVs), referred as "Large Oncosomes" (LO), have been identified in highly migratory and invasive PCa cells. We recently developed and characterized the DU145R80 subline, selected from parental DU145 cells as resistant to inhibitors of mevalonate pathway. DU145R80 showed different proteomic profile compared to parental DU145 cells, along with altered cytoskeleton dynamics and a more aggressive phenotype. METHODS Immunofluorescence staining and western blotting were used to identify blebbing and EVs protein cargo. EVs, purified by gradient ultra-centrifugations, were analyzed by tunable resistive pulse sensing and multi-parametric flow cytometry approach coupled with high-resolution imaging technologies. LO functional effects were tested in vitro by adhesion and invasion assays and in vivo xenograft model in nude mice. Xenograft and patient tumor tissues were analyzed by immunohistochemistry. RESULTS We found spontaneous blebbing and increased shedding of LO from DU145R80 compared to DU145 cells. LO from DU145R80, compared to those from DU145, carried increased amounts of key-molecules involved in PCa progression including integrin alpha V (αV-integrin). By incubating DU145 cells with DU145R80-derived LO we demonstrated that αV-integrin on LO surface was functionally involved in the increased adhesion and invasion of recipient cells, via AKT. Indeed either the pre-incubation of LO with an αV-integrin blocking antibody, or a specific AKT inhibition in recipient cells are able to revert the LO-induced functional effects. Moreover, DU145R80-derived LO also increased DU145 tumor engraftment in a mice model. Finally, we identified αV-integrin positive LO-like structures in tumor xenografts as well as in PCa patient tissues. Increased αV-integrin tumor expression correlated with high Gleason score and lymph node status. CONCLUSIONS Overall, this study is the first to demonstrate the critical role of αV-integrin positive LO in PCa aggressive features, adding new insights in biological function of these large EVs and suggesting their potential use as PCa prognostic markers.
Collapse
Affiliation(s)
- Chiara Ciardiello
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy.
| | - Alessandra Leone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Paola Lanuti
- Centre on Aging Sciences and Translational Medicine (Ce.S.I.-Me.T.), University "G.d'Annunzio", Chieti-Pescara, Italy.,Department of Medicine and Aging Sciences, University "G. d'Annunzio", Chieti-Pescara, Italy
| | - Maria S Roca
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Tania Moccia
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Valentina R Minciacchi
- Georg-Speyer-Haus Institute for Tumor biology and Experimental Therapy, Frankfurt, Germany
| | - Michele Minopoli
- Neoplastic Progression Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Naples, Italy
| | - Vincenzo Gigantino
- Pathology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Naples, Italy
| | - Rossella De Cecio
- Pathology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Naples, Italy
| | - Massimo Rippa
- Institute of Applied Sciences and Intelligent Systems 'E. Caianiello' of CNR, Pozzuoli, Italy
| | - Lucia Petti
- Institute of Applied Sciences and Intelligent Systems 'E. Caianiello' of CNR, Pozzuoli, Italy
| | - Francesca Capone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Carlo Vitagliano
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Maria R Milone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Biagio Pucci
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Rita Lombardi
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Federica Iannelli
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Francesca Bruzzese
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Marco Marchisio
- Centre on Aging Sciences and Translational Medicine (Ce.S.I.-Me.T.), University "G.d'Annunzio", Chieti-Pescara, Italy.,Department of Medicine and Aging Sciences, University "G. d'Annunzio", Chieti-Pescara, Italy
| | - Maria V Carriero
- Neoplastic Progression Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Naples, Italy
| | - Dolores Di Vizio
- Departments of Surgery, Pathology & Lab Medicine, and Biochemical Science, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy.
| |
Collapse
|
20
|
Sekino Y, Oue N, Mukai S, Shigematsu Y, Goto K, Sakamoto N, Sentani K, Hayashi T, Teishima J, Matsubara A, Yasui W. Protocadherin B9 promotes resistance to bicalutamide and is associated with the survival of prostate cancer patients. Prostate 2019; 79:234-242. [PMID: 30324761 DOI: 10.1002/pros.23728] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/27/2018] [Indexed: 12/20/2022]
Abstract
Background Prostate cancer (PCa) is a common malignancy worldwide and is the second leading cause of cancer death in men. The standard therapy for advanced PCa is androgen deprivation therapy (ADT). Although ADT, including bicalutamide treatment, is initially effective, resistance to bicalutamide frequently occurs and leads to the development of castration-resistant PCa. Thus, clarifying the mechanisms of bicalutamide resistance is urgently needed. We designed this study to assess the expression and function of PCDHB9, which encodes the protocadherin B9 protein. Methods The expression of PCDHB9 was determined using immunohistochemistry and a qRT-PCR. The effects of the overexpression or knockdown of PCDHB9 on cell growth, migration, adhesion were evaluated. To evaluate the PCDHB9-mediated effects in PCa, we performed a gene expression analysis using DU145 transfected with PCDHB9. We examined the effects of PCDHB9 inhibition on bicalutamide resistance. Results The qRT-PCR revealed that the expression of PCDHB9 was much higher in PCa than that in non-neoplastic prostate tissues. In 152 clinically localized PCa cases immunohistochemistry showed that 59% of PCa cases were positive for protocadherin B9. A Kaplan-Meier analysis showed that the high expression of protocadherin B9 was associated with PSA recurrence after radical prostatectomy. A functional analysis showed that PCDHB9 modulated cell migration and adhesion. We also found that PCDHB9 induced the expression of ITGB6 based on a gene expression analysis. The effect of PCDHB9 inhibition on bicalutamide sensitivity was examined using MTT assays. The IC50 value of PCDHB9 siRNA-transfected PCa cells was significantly lower than that of negative control siRNA-transfected cells. Furthermore, immunohistochemical staining of protocadherin B9 in 74 PCa patients who were treated with androgen depletion therapy, including bicalutamide treatment, demonstrated that the high expression of protocadherin B9 was significantly associated with poor overall survival. Conclusions PCDHB9 plays an important role in the progression of PCa and bicalutamide resistance. Collectively, our results suggest that PCDHB9 targeted therapy may be more effective than bicalutamide alone.
Collapse
Affiliation(s)
- Yohei Sekino
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Naohide Oue
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Shoichiro Mukai
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Graduate School of Biomedical and Health Sciences, Minami-ku, Hiroshima, Japan
| | - Yoshinori Shigematsu
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Keisuke Goto
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Naoya Sakamoto
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Kazuhiro Sentani
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Tetsutaro Hayashi
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Jun Teishima
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Akio Matsubara
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Wataru Yasui
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| |
Collapse
|
21
|
Nieberler M, Reuning U, Kessler H, Reichart F, Weirich G, Wolff KD. Fluorescence imaging of invasive head and neck carcinoma cells with integrin αvβ6-targeting RGD-peptides: an approach to a fluorescence-assisted intraoperative cytological assessment of bony resection margins. Br J Oral Maxillofac Surg 2018; 56:972-978. [PMID: 30502043 DOI: 10.1016/j.bjoms.2018.11.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 11/05/2018] [Indexed: 12/21/2022]
Abstract
We assessed the use of peptides containing arginylglycylaspartic acid (RGD) that target integrin αvβ6 as a potential approach for a fluorescence-assisted intraoperative cytological assessment of bony resection margins (F-AICAB) in patients who had bone-infiltrating squamous cell carcinoma (SCC) of the head and neck. This was assessed to demarcate invasive carcinoma cells that stained for αvβ6. Specimens from bony resection margins (n=362) were defined as either malignant or benign according to the results of cytological and histological examinations. Integrin αvβ6-targeting fluorescence-labelled RGD peptides were added to the cytological samples and the accuracy of the resulting signal assessed by comparing it with the cytological findings. The value of F-AICAB was evaluated to find out if it could help to improve future diagnoses, tests, and treatments. Integrin αvβ6 was strongly expressed in invasive SCC cells and qualified as a marker for bone-infiltrating carcinoma cells. It showed a high affinity to bind to invasive SCC cells and enabled swift and specific demarcation of αvβ6-stained carcinoma cells. It was also diagnostic, with a sensitivity of 100% (95% CI 81.3% to 99.3%), specificity of 98.3% (95% CI 94.4% to 99.0%), positive predictive value of 92% (95% CI 70.2% to 94.3%), and negative predictive value of 100% (95% CI 96.9% to 99.9%), compared with the cytological findings. The targeting of specific integrin subtypes with selective, synthetic ligands, adapted for multimodal imaging, is a promising new approach to diagnosis. Further studies are necessary to provide more evidence for successful clinical translation and to establish the impact on clinical procedures.
Collapse
Affiliation(s)
- M Nieberler
- Department of Oral and Maxillofacial Surgery, University Hospital rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81679 Munich, Germany.
| | - U Reuning
- Klinische Forschergruppe der Frauenklinik, University Hospital rechts der Isar, Technischen Universität München, Ismaninger Strasse 22, 81675 München, Germany
| | - H Kessler
- Institute for Advanced Study and Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany
| | - F Reichart
- Institute for Advanced Study and Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany
| | - G Weirich
- Institute of Pathology, Technische Universität München, Trogerstr. 18, 81675 Munich, Germany
| | - K-D Wolff
- Department of Oral and Maxillofacial Surgery, University Hospital rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81679 Munich, Germany
| |
Collapse
|
22
|
Lopatina T, Grange C, Fonsato V, Tapparo M, Brossa A, Fallo S, Pitino A, Herrera-Sanchez MB, Kholia S, Camussi G, Bussolati B. Extracellular vesicles from human liver stem cells inhibit tumor angiogenesis. Int J Cancer 2018; 144:322-333. [PMID: 30110127 DOI: 10.1002/ijc.31796] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 07/09/2018] [Accepted: 08/02/2018] [Indexed: 12/30/2022]
Abstract
Human liver stem-like cells (HLSC) and derived extracellular vesicles (EVs) were previously shown to exhibit anti-tumor activity. In our study, we investigated whether HLSC-derived EVs (HLSC-EVs) were able to inhibit tumor angiogenesis in vitro and in vivo, in comparison with EVs derived from mesenchymal stem cells (MSC-EVs). The results obtained indicated that HLSC-EVs, but not MSC-EVs, inhibited the angiogenic properties of tumor-derived endothelial cells (TEC) both in vitro and in vivo in a model of subcutaneous implantation in Matrigel. Treatment of TEC with HLSC-EVs led to the down-regulation of pro-angiogenic genes. Since HLSC-EVs carry a specific set of microRNAs (miRNAs) that could target these genes, we investigated their potential role by transfecting TEC with HLSC-EV specific miRNAs. We observed that four miRNAs, namely miR-15a, miR-181b, miR-320c and miR-874, significantly inhibited the angiogenic properties of TEC in vitro, and decreased the expression of some predicted target genes (ITGB3, FGF1, EPHB4 and PLAU). In parallel, TEC treated with HLSC-EVs significantly enhanced expression of miR-15a, miR-181b, miR-320c and miR-874 associated with the down-regulation of FGF1 and PLAU. In summary, HLSC-EVs possess an anti-tumorigenic effect, based on their ability to inhibit tumor angiogenesis.
Collapse
Affiliation(s)
- Tatiana Lopatina
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Cristina Grange
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Valentina Fonsato
- 2i3T, Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico, Scarl, University of Turin, Turin, Italy
| | - Marta Tapparo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alessia Brossa
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Sofia Fallo
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Adriana Pitino
- 2i3T, Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico, Scarl, University of Turin, Turin, Italy
| | - Maria Beatriz Herrera-Sanchez
- 2i3T, Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico, Scarl, University of Turin, Turin, Italy
| | - Sharad Kholia
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
23
|
Li X, Wang J, Pan Y, Xu Y, Liu D, Hou Y, Zhao G. Long non-coding RNA HULC affects the proliferation, apoptosis, migration, and invasion of mesenchymal stem cells. Exp Biol Med (Maywood) 2018; 243:1074-1082. [PMID: 30269516 DOI: 10.1177/1535370218804781] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
IMPACT STATEMENT Exploring the molecular mechanisms of growth and function in MSCs is the key to improve their clinical therapeutic effects. Currently, more and more evidence show that the long non-coding RNA (lncRNA) plays an important role in the growth, stemness and function of MSCs.Both HULC and MALAT1 are the earliest discovered LNCRNAs, which are closely related to tumor growth. All of them can promote the growth of liver cancer stem cells. Previously, we have studied the effects of MALAT1 on the growth and function of MSCs. In this study, we focused on the effects of HULC on MSCs. We elucidated the effects of HULC on the growth and differentiation of MSCs, and explored the relationship between inflammatory stimuli and HULC expression in MSCs. Our findings provide a new molecular target for the growth and clinical application of MSCs.
Collapse
Affiliation(s)
- Xiujun Li
- 1 Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jiali Wang
- 2 The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Yuchen Pan
- 1 Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yujun Xu
- 2 The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Dan Liu
- 1 Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yayi Hou
- 2 The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China.,3 Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Guangfeng Zhao
- 1 Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
24
|
Ozaki T, Yu M, Yin D, Sun D, Zhu Y, Bu Y, Sang M. Impact of RUNX2 on drug-resistant human pancreatic cancer cells with p53 mutations. BMC Cancer 2018; 18:309. [PMID: 29558908 PMCID: PMC5861661 DOI: 10.1186/s12885-018-4217-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 03/12/2018] [Indexed: 12/20/2022] Open
Abstract
Background Despite the remarkable advances in the early diagnosis and treatment, overall 5-year survival rate of patients with pancreatic cancer is less than 10%. Gemcitabine (GEM), a cytidine nucleoside analogue and ribonucleotide reductase inhibitor, is a primary option for patients with advanced pancreatic cancer; however, its clinical efficacy is extremely limited. This unfavorable clinical outcome of pancreatic cancer patients is at least in part attributable to their poor response to anti-cancer drugs such as GEM. Thus, it is urgent to understand the precise molecular basis behind the drug-resistant property of pancreatic cancer and also to develop a novel strategy to overcome this deadly disease. Review Accumulating evidence strongly suggests that p53 mutations contribute to the acquisition and/or maintenance of drug-resistant property of pancreatic cancer. Indeed, certain p53 mutants render pancreatic cancer cells much more resistant to GEM, implying that p53 mutation is one of the critical determinants of GEM sensitivity. Intriguingly, runt-related transcription factor 2 (RUNX2) is expressed at higher level in numerous human cancers such as pancreatic cancer and osteosarcoma, indicating that, in addition to its pro-osteogenic role, RUNX2 has a pro-oncogenic potential. Moreover, a growing body of evidence implies that a variety of miRNAs suppress malignant phenotypes of pancreatic cancer cells including drug resistance through the down-regulation of RUNX2. Recently, we have found for the first time that forced depletion of RUNX2 significantly increases GEM sensitivity of p53-null as well as p53-mutated pancreatic cancer cells through the stimulation of p53 family TAp63/TAp73-dependent cell death pathway. Conclusions Together, it is likely that RUNX2 is one of the promising molecular targets for the treatment of the patients with pancreatic cancer regardless of their p53 status. In this review article, we will discuss how to overcome the serious drug-resistant phenotype of pancreatic cancer.
Collapse
Affiliation(s)
- Toshinori Ozaki
- Laboratory of DNA Damage Signaling, Chiba Cancer Center Research Institute, Chiba, 260-8717, Japan.
| | - Meng Yu
- Department of Laboratory Animal of China Medical University, Shenyang, 110001, People's Republic of China
| | - Danjing Yin
- Research Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050017, People's Republic of China
| | - Dan Sun
- Department of Urology, First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Yuyan Zhu
- Department of Urology, First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Youquan Bu
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Meixiang Sang
- Research Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050017, People's Republic of China
| |
Collapse
|
25
|
Juan TK, Liu KC, Kuo CL, Yang MD, Chu YL, Yang JL, Wu PP, Huang YP, Lai KC, Chung JG. Tetrandrine suppresses adhesion, migration and invasion of human colon cancer SW620 cells via inhibition of nuclear factor-κB, matrix metalloproteinase-2 and matrix metalloproteinase-9 signaling pathways. Oncol Lett 2018; 15:7716-7724. [PMID: 29731901 PMCID: PMC5921181 DOI: 10.3892/ol.2018.8286] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 10/20/2017] [Indexed: 01/02/2023] Open
Abstract
Tetrandrine (TET) exhibits biological activities, including anticancer activity. In Chinese medicine, TET has been used to treat hypertensive and arrhythmic conditions and has been demonstrated to induce cytotoxic effects on human cancer cell lines. However, to the best of the author's knowledge, no previous studies have revealed that TET affects cell metastasis in SW620 human colon cancer cells. The present study demonstrated that TET decreased the cell number and inhibited cell adhesion and mobility of SW620 cells. Furthermore, a wound healing assay was performed to demonstrate that TET suppressed cell movement, and Transwell chamber assays were used to reveal that TET suppressed the cell migration and invasion of SW620 cells. Western blotting demonstrated that TET significantly reduced protein expression levels of SOS Ras/Rac guanine nucleotide exchange factor 1, phosphatidylinositol 3-kinase, growth factor receptor bound protein 2, phosphorylated (p)-c Jun N-terminal kinase 1/2, p-p38, p38, 14-3-3, Rho A, β-catenin, nuclear factor-κB p65, signal transducer and activator of transcription-1 and cyclooxygenase-2, in comparison with untreated SW620 cells. Overall, the results of the present study suggested that TET may be used as a novel anti-metastasis agent for the treatment of human colon cancer in the future.
Collapse
Affiliation(s)
- Ta-Kuo Juan
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Kuo-Ching Liu
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Chao-Lin Kuo
- Chinese Medicine Resources, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Mei-Due Yang
- Department of Surgery, China Medical University Hospital, Taichung 404, Taiwan, R.O.C
| | - Yung-Lin Chu
- International Master's Degree Program in Food Science, International College, National Pingtung University of Science and Technology, Pingtung 912, Taiwan, R.O.C
| | - Jiun-Long Yang
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Ping-Ping Wu
- School of Pharmacy, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Yi-Ping Huang
- Department of Physiology, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Kuang-Chi Lai
- School of Medicine, China Medical University, Taichung 404, Taiwan, R.O.C.,Department of Medical Laboratory Science and Biotechnology, College of Medicine and Life Science, Chung Hwa University of Medical Technology, Tainan 717, Taiwan, R.O.C.,Department of Surgery, China Medical University Beigang Hospital, Beigang, Yunlin 651, Taiwan, R.O.C
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan, R.O.C.,Department of Biotechnology, Asia University, Wufeng, Taichung 413, Taiwan, R.O.C
| |
Collapse
|
26
|
Lu H, Bowler N, Harshyne LA, Craig Hooper D, Krishn SR, Kurtoglu S, Fedele C, Liu Q, Tang HY, Kossenkov AV, Kelly WK, Wang K, Kean RB, Weinreb PH, Yu L, Dutta A, Fortina P, Ertel A, Stanczak M, Forsberg F, Gabrilovich DI, Speicher DW, Altieri DC, Languino LR. Exosomal αvβ6 integrin is required for monocyte M2 polarization in prostate cancer. Matrix Biol 2018. [PMID: 29530483 DOI: 10.1016/j.matbio.2018.03.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Therapeutic approaches aimed at curing prostate cancer are only partially successful given the occurrence of highly metastatic resistant phenotypes that frequently develop in response to therapies. Recently, we have described αvβ6, a surface receptor of the integrin family as a novel therapeutic target for prostate cancer; this epithelial-specific molecule is an ideal target since, unlike other integrins, it is found in different types of cancer but not in normal tissues. We describe a novel αvβ6-mediated signaling pathway that has profound effects on the microenvironment. We show that αvβ6 is transferred from cancer cells to monocytes, including β6-null monocytes, by exosomes and that monocytes from prostate cancer patients, but not from healthy volunteers, express αvβ6. Cancer cell exosomes, purified via density gradients, promote M2 polarization, whereas αvβ6 down-regulation in exosomes inhibits M2 polarization in recipient monocytes. Also, as evaluated by our proteomic analysis, αvβ6 down-regulation causes a significant increase in donor cancer cells, and their exosomes, of two molecules that have a tumor suppressive role, STAT1 and MX1/2. Finally, using the Ptenpc-/- prostate cancer mouse model, which carries a prostate epithelial-specific Pten deletion, we demonstrate that αvβ6 inhibition in vivo causes up-regulation of STAT1 in cancer cells. Our results provide evidence of a novel mechanism that regulates M2 polarization and prostate cancer progression through transfer of αvβ6 from cancer cells to monocytes through exosomes.
Collapse
Affiliation(s)
- Huimin Lu
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Nicholas Bowler
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Larry A Harshyne
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - D Craig Hooper
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Shiv Ram Krishn
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Senem Kurtoglu
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Carmine Fedele
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Qin Liu
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA, USA
| | - Hsin-Yao Tang
- Center for Systems and Computational Biology, Wistar Institute, Philadelphia, PA, USA
| | - Andrew V Kossenkov
- Center for Systems and Computational Biology, Wistar Institute, Philadelphia, PA, USA
| | - William K Kelly
- Departments of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kerith Wang
- Departments of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rhonda B Kean
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Lei Yu
- Flow Cytometry Core Facility, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Anindita Dutta
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Paolo Fortina
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Cancer Genomics and Bioinformatics Laboratory, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam Ertel
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Cancer Genomics and Bioinformatics Laboratory, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Maria Stanczak
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dmitry I Gabrilovich
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Immunology, Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA, USA
| | - David W Speicher
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA, USA; Center for Systems and Computational Biology, Wistar Institute, Philadelphia, PA, USA
| | - Dario C Altieri
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Immunology, Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA, USA
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
27
|
Farina NH, Zingiryan A, Akech JA, Callahan CJ, Lu H, Stein JL, Languino LR, Stein GS, Lian JB. A microRNA/Runx1/Runx2 network regulates prostate tumor progression from onset to adenocarcinoma in TRAMP mice. Oncotarget 2018; 7:70462-70474. [PMID: 27634876 PMCID: PMC5342565 DOI: 10.18632/oncotarget.11992] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/02/2016] [Indexed: 01/08/2023] Open
Abstract
While decades of research have identified molecular pathways inducing and promoting stages of prostate cancer malignancy, studies addressing dynamic changes of cancer-related regulatory factors in a prostate tumor progression model are limited. Using the TRAMP mouse model of human prostate cancer, we address mechanisms of deregulation for the cancer-associated transcription factors, Runx1 and Runx2 by identifying microRNAs with reciprocal expression changes at six time points during 33 weeks of tumorigenesis. We molecularly define transition stages from PIN lesions to hyperplasia/neoplasia and progression to adenocarcinoma by temporal changes in expression of human prostate cancer markers, including the androgen receptor and tumor suppressors, Nkx3.1 and PTEN. Concomitant activation of PTEN, AR, and Runx factors occurs at early stages. At late stages, PTEN and AR are downregulated, while Runx1 and Runx2 remain elevated. Loss of Runx-targeting microRNAs, miR-23b-5p, miR-139-5p, miR-205-5p, miR-221-3p, miR-375-3p, miR-382-5p, and miR-384-5p, contribute to aberrant Runx expression in prostate tumors. Our studies reveal a Runx/miRNA interaction axis centered on PTEN-PI3K-AKT signaling. This regulatory network translates to mechanistic understanding of prostate tumorigenesis that can be developed for diagnosis and directed therapy.
Collapse
Affiliation(s)
- Nicholas H Farina
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | - Areg Zingiryan
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | - Jacqueline A Akech
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Cody J Callahan
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | - Huimin Lu
- Prostate Cancer Discovery and Development Program, Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Janet L Stein
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Gary S Stein
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | - Jane B Lian
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT 05405, USA
| |
Collapse
|
28
|
Kubyshkin AV, Fomochkina II, Petrosyan AM. THE IMPACT OF ALCOHOL ON PRO-METASTATIC N-GLYCOSYLATION IN PROSTATE CANCER. KRIMSKII ZHURNAL EKSPERIMENTAL'NOI I KLINICHESKOI MEDITSINY = KRYMS'KYI ZHURNAL EKSPERYMENTAL'NOI TA KLINICHNOI MEDYTSYNY = CRIMEAN JOURNAL OF EXPERIMENTAL AND CLINICAL MEDICINE 2018; 8:11-20. [PMID: 31131224 PMCID: PMC6534161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Chronic alcohol abuse and alcoholism are considered risk factors for prostate cancer (PCa) progression, but the mechanism is unknown. Previously, we found that: (1) fragmentation of the Golgi complex correlates with the progression of PCa; (2) ethanol (EtOH) induces Golgi disorganization, which, in turn, alters intra-Golgi localization of some Golgi proteins. Also, progression of the prostate tumor is associated with activation of N-acetylglucosaminyltransferase-V (MGAT5)-mediated N-glycosylation of pro-metastatic proteins, including matriptase and integrins, followed by their enhanced retention at the cell surface. Here, using high-resolution microscopy, we found that alcohol effect on Golgi in low passage androgen-responsive LNCaP cells mimic the fragmented Golgi phenotype of androgen-refractory high passage LNCaP and PC-3 cells. Next, we detected that transition to androgen unresponsiveness is accompanied by downregulation of N-acetylglucosaminyltransferase-III (MGAT3), the enzyme that competes with MGAT5 for anti-metastatic N-glycan branching. Moreover, in low passage LNCaP cells, alcohol-induced Golgi fragmentation induced translocation of MGAT3 from the Golgi to the cytoplasm, while intra-Golgi localization of MGAT5 appeared unaffected. Then, the relationship between Golgi morphology, MGAT3 intracellular position, and clinicopathologic features was assessed in human PCa patient specimens with and without a history of alcohol dependence. We revealed that within the same clinical stage, the level of Golgi disorganization and the cytoplasmic shift of MGAT3 was more prominent in patients consuming alcohol. In vitro studies suggest that EtOH-induced downregulation of MGAT3 correlates with activation of MGAT5-mediated glycosylation and overexpression of both matriptase and integrins. In sum, we provide a novel insight into the alcohol-mediated tumor promotion.
Collapse
Affiliation(s)
- A V Kubyshkin
- Medical Academy named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University, Lenin Avenue 5/7, Simferopol, Russia; 295051
| | - I I Fomochkina
- Medical Academy named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University, Lenin Avenue 5/7, Simferopol, Russia; 295051
| | - A M Petrosyan
- University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE, USA; 68198-5870
| |
Collapse
|
29
|
Li Y, Chen Y, Ma Y, Nenkov M, Haase D, Petersen I. Collagen prolyl hydroxylase 3 has a tumor suppressive activity in human lung cancer. Exp Cell Res 2017; 363:121-128. [PMID: 29277505 DOI: 10.1016/j.yexcr.2017.12.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/18/2017] [Accepted: 12/21/2017] [Indexed: 11/27/2022]
Abstract
Collagen prolyl hydroxylases (P3H) are required for proper collagen biosynthesis. One of the family members P3H3 was downregulated in breast cancer and lymphoma due to DNA methylation. However the role of P3H3 in lung cancer has not yet been elucidated. In this study, we analyzed P3H3 expression in a panel of lung cancer cell lines and primary lung tumors. Epigenetic regulation was explored and the function of P3H3 was investigated by stable transfection and RNA interference. We found that P3H3 was downregulated in 6 out of 10 lung cancer cell lines. A heterogeneous methylation pattern of P3H3 was found in the exon region. In primary lung tumors, immunohistochemistry on tissue microarray (TMA) showed that higher expression of P3H3 was significantly associated with lower tumor N stage and grade (p = 0.035 and p = 0.026, respectively). Ectopic expression of P3H3 inhibited cell proliferation, colony formation, migration as well as invasion, and induced apoptosis together with cell cycle arrest in the G2/M phase. Knockdown of P3H3 led to increased migratory and invasive potential. These Phenomena are accompanied by enhanced p21, decreased cyclin A1 levels and increased caspase 3/7 activities. Taken together, we feel that P3H3 is a novel tumor suppressor and its protein expression is inversely related to lymph node metastasis and tumor differentiation in lung cancer.
Collapse
Affiliation(s)
- Yong Li
- Institute of Pathology, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Yuan Chen
- Institute of Pathology, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Yunxia Ma
- Institute of Pathology, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Miljana Nenkov
- Institute of Pathology, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany.
| | - Daniela Haase
- Institute of Pathology, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Iver Petersen
- Institute of Pathology, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
30
|
Uusi-Kerttula H, Davies J, Coughlan L, Hulin-Curtis S, Jones R, Hanna L, Chester JD, Parker AL. Pseudotyped αvβ6 integrin-targeted adenovirus vectors for ovarian cancer therapies. Oncotarget 2017; 7:27926-37. [PMID: 27056886 PMCID: PMC5053699 DOI: 10.18632/oncotarget.8545] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/28/2016] [Indexed: 01/02/2023] Open
Abstract
Encouraging results from recent clinical trials are revitalizing the field of oncolytic virotherapies. Human adenovirus type 5 (HAdV-C5/Ad5) is a common vector for its ease of manipulation, high production titers and capacity to transduce multiple cell types. However, effective clinical applications are hindered by poor tumor-selectivity and vector neutralization. We generated Ad5/kn48 by pseudotyping Ad5 with the fiber knob domain from the less seroprevalent HAdV-D48 (Ad48). The vector was shown to utilize coxsackie and adenovirus receptor (CAR) but not CD46 for cell entry. A 20-amino acid peptide NAVPNLRGDLQVLAQKVART (A20) was inserted into the Ad5. Luc HI loop (Ad5.HI.A20) and Ad5/kn48 DG loop (Ad5/kn48.DG.A20) to target a prognostic cancer cell marker, αvβ6 integrin. Relative to the Ad5.Luc parent vector, Ad5.HI.A20, Ad5.KO1.HI.A20 (KO1, ablated CAR-binding) and Ad5/kn48.DG.A20 showed ~ 160-, 270- and 180-fold increased transduction in BT-20 breast carcinoma cells (αvβ6high). Primary human epithelial ovarian cancer (EOC) cultures derived from clinical ascites provided a useful ex vivo model for intraperitoneal virotherapy. Ad5.HI.A20, Ad5.KO1.HI.A20 and Ad5/kn48.DG.A20 transduction was ~ 70-, 60- and 16-fold increased relative to Ad5.Luc in EOC cells (αvβ6high), respectively. A20 vectors transduced EOC cells at up to ~ 950-fold higher efficiency in the presence of neutralizing ovarian ascites, as compared to Ad5.Luc. Efficient transduction and enhanced cancer-selectivity via a non-native αvβ6-mediated route was demonstrated, even in the presence of pre-existing anti-Ad5 immunity. Consequently, αvβ6-targeted Ad vectors may represent a promising platform for local intraperitoneal treatment of ovarian cancer metastases.
Collapse
Affiliation(s)
- Hanni Uusi-Kerttula
- Department of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - James Davies
- Department of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Lynda Coughlan
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Sarah Hulin-Curtis
- Department of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | | | | | - John D Chester
- Department of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK.,Velindre Cancer Centre, Cardiff CF14 2TL, UK
| | - Alan L Parker
- Department of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| |
Collapse
|
31
|
Zhang C, Winnard PT, Dasari S, Kominsky SL, Doucet M, Jayaraman S, Raman V, Barman I. Label-free Raman spectroscopy provides early determination and precise localization of breast cancer-colonized bone alterations. Chem Sci 2017; 9:743-753. [PMID: 29629144 PMCID: PMC5869989 DOI: 10.1039/c7sc02905e] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 11/14/2017] [Indexed: 12/13/2022] Open
Abstract
Raman spectral markers offer new routes to recognition of biomolecular alterations at sites of nascent and progressing metastatic cancer in bone.
Breast neoplasms frequently colonize bone and induce development of osteolytic bone lesions by disrupting the homeostasis of the bone microenvironment. This degenerative process can lead to bone pain and pathological bone fracture, a major cause of cancer morbidity and diminished quality of life, which is exacerbated by our limited ability to monitor early metastatic disease in bone and assess fracture risk. Spurred by its label-free, real-time nature and its exquisite molecular specificity, we employed spontaneous Raman spectroscopy to assess and quantify early metastasis driven biochemical alterations to bone composition. As early as two weeks after intracardiac inoculations of MDA-MB-435 breast cancer cells in NOD-SCID mice, Raman spectroscopic measurements in the femur and spine revealed consistent changes in carbonate substitution, overall mineralization as well as crystallinity increase in tumor-bearing bones when compared with their normal counterparts. Our observations reveal the possibility of early stage detection of biochemical changes in the tumor-bearing bones – significantly before morphological variations are captured through radiographic diagnosis. This study paves the way for a better molecular understanding of altered bone remodeling in such metastatic niches, and for further clinical studies with the goal of establishing a non-invasive tool for early metastasis detection and prediction of pathological fracture risk in breast cancer.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Mechanical Engineering , Johns Hopkins University , Whiting School of Engineering , Latrobe Hall 103 , Baltimore , MD 21218 , USA . ; Tel: +1-410-516-0656
| | - Paul T Winnard
- Division of Cancer Imaging Research , Russell H. Morgan Department of Radiology and Radiological Science , Johns Hopkins University School of Medicine , 720 Rutland Avenue, Rm 340 Traylor Building , Baltimore , MD , USA 21205 . ; Tel: +1-410-955-7492
| | - Sidarth Dasari
- Indiana University School of Medicine , Indianapolis , IN , USA
| | - Scott L Kominsky
- Department of Orthopaedic Surgery , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Michele Doucet
- Department of Orthopaedic Surgery , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Swaathi Jayaraman
- Department of Orthopaedic Surgery , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Venu Raman
- Division of Cancer Imaging Research , Russell H. Morgan Department of Radiology and Radiological Science , Johns Hopkins University School of Medicine , 720 Rutland Avenue, Rm 340 Traylor Building , Baltimore , MD , USA 21205 . ; Tel: +1-410-955-7492.,Department of Oncology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Ishan Barman
- Department of Mechanical Engineering , Johns Hopkins University , Whiting School of Engineering , Latrobe Hall 103 , Baltimore , MD 21218 , USA . ; Tel: +1-410-516-0656.,Department of Oncology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| |
Collapse
|
32
|
Abstract
Transforming growth factor βs (TGF-βs) are closely related ligands that have pleiotropic activity on most cell types of the body. They act through common heterotetrameric TGF-β type II and type I transmembrane dual specificity kinase receptor complexes, and the outcome of signaling is context-dependent. In normal tissue, they serve a role in maintaining homeostasis. In many diseased states, particularly fibrosis and cancer, TGF-β ligands are overexpressed and the outcome of signaling is diverted toward disease progression. There has therefore been a concerted effort to develop drugs that block TGF-β signaling for therapeutic benefit. This review will cover the basics of TGF-β signaling and its biological activities relevant to oncology, present a summary of pharmacological TGF-β blockade strategies, and give an update on preclinical and clinical trials for TGF-β blockade in a variety of solid tumor types.
Collapse
Affiliation(s)
- Rosemary J Akhurst
- Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94158-9001
| |
Collapse
|
33
|
Abstract
Transforming growth factor βs (TGF-βs) are closely related ligands that have pleiotropic activity on most cell types of the body. They act through common heterotetrameric TGF-β type II and type I transmembrane dual specificity kinase receptor complexes, and the outcome of signaling is context-dependent. In normal tissue, they serve a role in maintaining homeostasis. In many diseased states, particularly fibrosis and cancer, TGF-β ligands are overexpressed and the outcome of signaling is diverted toward disease progression. There has therefore been a concerted effort to develop drugs that block TGF-β signaling for therapeutic benefit. This review will cover the basics of TGF-β signaling and its biological activities relevant to oncology, present a summary of pharmacological TGF-β blockade strategies, and give an update on preclinical and clinical trials for TGF-β blockade in a variety of solid tumor types.
Collapse
Affiliation(s)
- Rosemary J Akhurst
- Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94158-9001
| |
Collapse
|
34
|
Paolillo M, Schinelli S. Integrins and Exosomes, a Dangerous Liaison in Cancer Progression. Cancers (Basel) 2017; 9:cancers9080095. [PMID: 28933725 PMCID: PMC5575598 DOI: 10.3390/cancers9080095] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/20/2017] [Accepted: 07/22/2017] [Indexed: 12/19/2022] Open
Abstract
Integrin activity and function is classically related to the bi-directional regulation of cell-extracellular matrix (ECM) contacts that regulate a number of cell pathways linked to cell adhesion, cell detachment from ECM, cell migration, and anoikis. Interestingly, emerging data continue to uncover new roles for integrins in cancer-relevant pathways, particularly concerning the regulation of immune cell activity in the tumor niche, like myeloid cell differentiation and function and, very recently, the regulation of metastatic processes by exosomes. Exosomes are deeply involved in cell-cell communication processes and several studies have shown that integrins found in tumor-associated exosomes can promote cancer progression by two novel cooperative mechanisms: horizontal transfer of integrin transcripts as vescicle cargo, and selection of target tissues to form new tumor niches during metastatic spread by integrins carried on the exosome’s surface. In this review we will discuss mounting evidence that contribute to the development of a new picture for integrins in cancer, highlighting the role of integrins in the processes that leads to tumor niche formation. In particular, the role of the periostin pathway in the recruitment of tumor-associated macrophages, and the proposed contribution of exosome-derived integrins in the metastatic spread will be discussed. Finally, in light of the above considerations, an evaluation of integrins as possible therapeutic targets will be conducted.
Collapse
Affiliation(s)
- Mayra Paolillo
- Department of Drug Sciences, University of Pavia, Viale Taramelli 14, Pavia 27100, Italy.
| | - Sergio Schinelli
- Department of Drug Sciences, University of Pavia, Viale Taramelli 14, Pavia 27100, Italy.
| |
Collapse
|
35
|
The traditional Chinese medicine Achyranthes bidentata and our de novo conception of its metastatic chemoprevention: from phytochemistry to pharmacology. Sci Rep 2017. [PMID: 28634392 PMCID: PMC5478643 DOI: 10.1038/s41598-017-02054-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Our recent biosystems analysis revealed similarities between embryonic implantation and cancer cell adhesion, which suggests that abortifacients may be good for safe and effective metastatic chemoprevention targeting circulating tumor cells (CTC). Here we test the hypothesis by using the well-known abortion herb Achyranthes bidentata Blume (A. bidentata). Five compounds were separated from the herb root. Among them, ginsenoside Ro was the most potent in inhibiting embryonic implantation within non-cytotoxic concentrations. It specifically inhibited the metastatic dissemination capability of colon cancer cells HT29, including the migration and invasion ability, and their adhesion to human endothelium through inhibiting integrin αvβ6, MMP-2, MMP-9, and ERK phosphorylation by HT29. Pretreatment of nude mice with oral ginsenoside Ro followed by HT29 intravenous inoculation and 40-day oral ginsenoside Ro significantly prevented lung metastasis with downregulation of integrin αvβ6 and no toxicity. The present study firstly introduces the new conception of utilizing safe and effective abortion botanic medicines for CTC-based metastatic chemoprevention.
Collapse
|
36
|
Asgharzadeh MR, Barar J, Pourseif MM, Eskandani M, Jafari Niya M, Mashayekhi MR, Omidi Y. Molecular machineries of pH dysregulation in tumor microenvironment: potential targets for cancer therapy. BIOIMPACTS : BI 2017; 7:115-133. [PMID: 28752076 PMCID: PMC5524986 DOI: 10.15171/bi.2017.15] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/28/2017] [Accepted: 06/06/2017] [Indexed: 12/30/2022]
Abstract
Introduction: Cancer is an intricate disorder/dysfunction of cells that can be defined as a genetic heterogeneity in human disease. Therefore, it is characterized by several adaptive complex hallmarks. Among them, the pH dysregulation appears as a symbol of aberrant functions within the tumor microenvironment (TME). In comparison with normal tissues, in the solid tumors, we face with an irregular acidification and alkalinization of the extracellular and intracellular fluids. Methods: In this study, we comprehensively discussed the most recent reports on the hallmarks of solid tumors to provide deep insights upon the molecular machineries involved in the pH dysregulation of solid tumors and their impacts on the initiation and progression of cancer. Results: The dysregulation of pH in solid tumors is fundamentally related to the Warburg effect and hypoxia, leading to expression of a number of molecular machineries, including: NHE1, H+ pump V-ATPase, CA-9, CA-12, MCT-1, GLUT-1. Activation of proton exchangers and transporters (PETs) gives rise to formation of TME. This condition favors the cancer cells to evade from the anoikis and apoptosis, granting them aggressive and metastasis phenotype, as well as resistance to chemotherapy and radiation therapy. This review aimed to discuss the key molecular changes of tumor cells in terms of bio-energetics and cancer metabolism in relation with pH dysregulation. During this phenomenon, the intra- and extracellular metabolites are altered and/or disrupted. Such molecular alterations provide molecular hallmarks for direct targeting of the PETs by potent relevant inhibitors in combination with conventional cancer therapies as ultimate therapy against solid tumors. Conclusion: Taken all, along with other treatment strategies, targeting the key molecular machineries related to intra- and extracellular metabolisms within the TME is proposed as a novel strategy to inhibit or block PETs that are involved in the pH dysregulation of solid tumors.
Collapse
Affiliation(s)
- Mohammad Reza Asgharzadeh
- Department of Biology, Fars Science and Research Branch, Islamic Azad University, Marvdasht, Iran
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad M. Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojtaba Jafari Niya
- Department of Biology, Fars Science and Research Branch, Islamic Azad University, Marvdasht, Iran
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | | | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
37
|
Ahmedah HT, Patterson LH, Shnyder SD, Sheldrake HM. RGD-Binding Integrins in Head and Neck Cancers. Cancers (Basel) 2017; 9:cancers9060056. [PMID: 28587135 PMCID: PMC5483875 DOI: 10.3390/cancers9060056] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 12/13/2022] Open
Abstract
Alterations in integrin expression and function promote tumour growth, invasion, metastasis and neoangiogenesis. Head and neck cancers are highly vascular tumours with a tendency to metastasise. They express a wide range of integrin receptors. Expression of the αv and β1 subunits has been explored relatively extensively and linked to tumour progression and metastasis. Individual receptors αvβ3 and αvβ5 have proved popular targets for diagnostic and therapeutic agents but lesser studied receptors, such as αvβ6, αvβ8, and β1 subfamily members, also show promise. This review presents the current knowledge of integrin expression and function in squamous cell carcinoma of the head and neck (HNSCC), with a particular focus on the arginine-glycine-aspartate (RGD)-binding integrins, in order to highlight the potential of integrins as targets for personalised tumour-specific identification and therapy.
Collapse
Affiliation(s)
- Hanadi Talal Ahmedah
- Radiological Sciences Department, College of Health and Rehabilitation Sciences, Princess Nourah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia.
| | | | - Steven D Shnyder
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK.
| | - Helen M Sheldrake
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK.
| |
Collapse
|
38
|
Chen PC, Tang CH, Lin LW, Tsai CH, Chu CY, Lin TH, Huang YL. Thrombospondin-2 promotes prostate cancer bone metastasis by the up-regulation of matrix metalloproteinase-2 through down-regulating miR-376c expression. J Hematol Oncol 2017; 10:33. [PMID: 28122633 PMCID: PMC5264454 DOI: 10.1186/s13045-017-0390-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 01/03/2017] [Indexed: 11/24/2022] Open
Abstract
Background Thrombospondin-2 (TSP-2) is a secreted matricellular glycoprotein that is found to mediate cell-to-extracellular matrix attachment and participates in many physiological and pathological processes. The expression profile of TSP-2 on tumors is controversial, and it up-regulates in some cancers, whereas it down-regulates in others, suggesting that the functional role of TSP-2 on tumors is still uncertain. Methods The expression of TSP-2 on prostate cancer progression was determined in the tissue array by the immunohistochemistry. The molecular mechanism of TSP-2 on prostate cancer (PCa) metastasis was investigated through pharmaceutical inhibitors, siRNAs, and miRNAs analyses. The role of TSP-2 on PCa metastasis in vivo was verified through xenograft in vivo imaging system. Results Based on the gene expression omnibus database and immunohistochemistry, we found that TSP-2 increased with the progression of PCa, especially in metastatic PCa and is correlated with the matrix metalloproteinase-2 (MMP-2) expression. Additionally, through binding to CD36 and integrin ανβ3, TSP-2 increased cell migration and MMP-2 expression. With inhibition of p38, ERK, and JNK, the TSP-2-induced cell migration and MMP-2 expression were abolished, indicating that the TSP-2’s effect on PCa is MAPK dependent. Moreover, the microRNA-376c (miR-376c) was significantly decreased by the TSP-2 treatment. Furthermore, the TSP-2-induced MMP-2 expression and the subsequent cell motility were suppressed upon miR-376c mimic stimulation. On the other hand, the animal studies revealed that the bone metastasis was abolished when TSP-2 was stably knocked down in PCa cells. Conclusions Taken together, our results indicate that TSP-2 enhances the migration of PCa cells by increasing MMP-2 expression through down-regulation of miR-376c expression. Therefore, TSP-2 may represent a promising new target for treating PCa. Electronic supplementary material The online version of this article (doi:10.1186/s13045-017-0390-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Po-Chun Chen
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Pharmacology, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Liang-Wei Lin
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Orthopedic Surgery, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Cheng-Ying Chu
- The Ph.D. Program for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Tien-Huang Lin
- Department of Urology, Buddhist Tzu Chi General Hospital Taichung Branch, Taichung, Taiwan
| | - Yuan-Li Huang
- Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan. .,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
39
|
Tauro M, Shay G, Sansil SS, Laghezza A, Tortorella P, Neuger AM, Soliman H, Lynch CC. Bone-Seeking Matrix Metalloproteinase-2 Inhibitors Prevent Bone Metastatic Breast Cancer Growth. Mol Cancer Ther 2017; 16:494-505. [PMID: 28069877 DOI: 10.1158/1535-7163.mct-16-0315-t] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 12/05/2016] [Accepted: 12/07/2016] [Indexed: 11/16/2022]
Abstract
Bone metastasis is common during breast cancer progression. Matrix metalloproteinase-2 (MMP-2) is significantly associated with aggressive breast cancer and poorer overall survival. In bone, tumor- or host-derived MMP-2 contributes to breast cancer growth and does so by processing substrates, including type I collagen and TGFβ latency proteins. These data provide strong rationale for the application of MMP-2 inhibitors to treat the disease. However, in vivo, MMP-2 is systemically expressed. Therefore, to overcome potential toxicities noted with previous broad-spectrum MMP inhibitors (MMPIs), we used highly selective bisphosphonic-based MMP-2 inhibitors (BMMPIs) that allowed for specific bone targeting. In vitro, BMMPIs affected the viability of breast cancer cell lines and osteoclast precursors, but not osteoblasts. In vivo, we demonstrated using two bone metastatic models (PyMT-R221A and 4T1) that BMMPI treatment significantly reduced tumor growth and tumor-associated bone destruction. In addition, BMMPIs are superior in promoting tumor apoptosis compared with the standard-of-care bisphosphonate, zoledronate. We demonstrated MMP-2-selective inhibition in the bone microenvironment using specific and broad-spectrum MMP probes. Furthermore, compared with zoledronate, BMMPI-treated mice had significantly lower levels of TGFβ signaling and MMP-generated type I collagen carboxy-terminal fragments. Taken together, our data show the feasibility of selective inhibition of MMPs in the bone metastatic breast cancer microenvironment. We posit that BMMPIs could be easily translated to the clinical setting for the treatment of bone metastases given the well-tolerated nature of bisphosphonates. Mol Cancer Ther; 16(3); 494-505. ©2017 AACR.
Collapse
Affiliation(s)
- Marilena Tauro
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Gemma Shay
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Samer S Sansil
- Translational Research Core and, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Antonio Laghezza
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari "A. Moro", Bari, Italy
| | - Paolo Tortorella
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari "A. Moro", Bari, Italy
| | - Anthony M Neuger
- Translational Research Core and, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Hatem Soliman
- Department of Women's Oncology and Experimental Therapeutics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Conor C Lynch
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| |
Collapse
|
40
|
Bianconi D, Unseld M, Prager GW. Integrins in the Spotlight of Cancer. Int J Mol Sci 2016; 17:ijms17122037. [PMID: 27929432 PMCID: PMC5187837 DOI: 10.3390/ijms17122037] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/17/2016] [Accepted: 11/28/2016] [Indexed: 02/07/2023] Open
Abstract
Integrins are heterodimeric cell surface receptors that bind to different extracellular ligands depending on their composition and regulate all processes which enable multicellular life. In cancer, integrins trigger and play key roles in all the features that were once described as the Hallmarks of Cancer. In this review, we will discuss the contribution of integrins to these hallmarks, including uncontrolled and limitless proliferation, invasion of tumor cells, promotion of tumor angiogenesis and evasion of apoptosis and resistance to growth suppressors, by highlighting the latest findings. Further on, given the paramount role of integrins in cancer, we will present novel strategies for integrin inhibition that are starting to emerge, promising a hopeful future regarding cancer treatment.
Collapse
Affiliation(s)
- Daniela Bianconi
- Department of Internal Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Matthias Unseld
- Department of Internal Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Gerald W Prager
- Department of Internal Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, A-1090 Vienna, Austria.
| |
Collapse
|
41
|
Lu H, Wang T, Li J, Fedele C, Liu Q, Zhang J, Jiang Z, Jain D, Iozzo RV, Violette SM, Weinreb PH, Davis RJ, Gioeli D, FitzGerald TJ, Altieri DC, Languino LR. αvβ6 Integrin Promotes Castrate-Resistant Prostate Cancer through JNK1-Mediated Activation of Androgen Receptor. Cancer Res 2016; 76:5163-74. [PMID: 27450452 DOI: 10.1158/0008-5472.can-16-0543] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 06/20/2016] [Indexed: 12/20/2022]
Abstract
Androgen receptor signaling fuels prostate cancer and is a major therapeutic target. However, mechanisms of resistance to therapeutic androgen ablation are not well understood. Here, using a prostate cancer mouse model, Pten(pc-/-), carrying a prostate epithelial-specific Pten deletion, we show that the αvβ6 integrin is required for tumor growth in vivo of castrated as well as of noncastrated mice. We describe a novel signaling pathway that couples the αvβ6 integrin cell surface receptor to androgen receptor via activation of JNK1 and causes increased nuclear localization and activity of androgen receptor. This downstream kinase activation by αvβ6 is specific for JNK1, with no involvement of p38 or ERK kinase. In addition, differential phosphorylation of Akt is not observed under these conditions, nor is cell morphology affected by αvβ6 expression. This pathway, which is specific for αvβ6, because it is not regulated by a different αv-containing integrin, αvβ3, promotes upregulation of survivin, which in turn supports anchorage-independent growth of αvβ6-expressing cells. Consistently, both αvβ6 and survivin are significantly increased in prostatic adenocarcinoma, but are not detected in normal prostatic epithelium. Neither XIAP nor Bcl-2 is affected by αvβ6 expression. In conclusion, we show that αvβ6 expression is required for prostate cancer progression, including castrate-resistant prostate cancer; mechanistically, by promoting activation of JNK1, the αvβ6 integrin causes androgen receptor-increased activity in the absence of androgen and consequent upregulation of survivin. These preclinical results pave the way for further clinical development of αvβ6 antagonists for prostate cancer therapy. Cancer Res; 76(17); 5163-74. ©2016 AACR.
Collapse
Affiliation(s)
- Huimin Lu
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Tao Wang
- Department of Radiation Oncology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Jing Li
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Carmine Fedele
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Qin Liu
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania. Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Jianzhong Zhang
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Zhong Jiang
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Dhanpat Jain
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | | | - Roger J Davis
- Program in Molecular Medicine and Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Daniel Gioeli
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Thomas J FitzGerald
- Department of Radiation Oncology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Dario C Altieri
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania. Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
42
|
Singh A, Fedele C, Lu H, Nevalainen MT, Keen JH, Languino LR. Exosome-mediated Transfer of αvβ3 Integrin from Tumorigenic to Nontumorigenic Cells Promotes a Migratory Phenotype. Mol Cancer Res 2016; 14:1136-1146. [PMID: 27439335 DOI: 10.1158/1541-7786.mcr-16-0058] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 06/13/2016] [Accepted: 07/03/2016] [Indexed: 12/20/2022]
Abstract
The αvβ3 integrin is known to be highly upregulated during cancer progression and promotes a migratory and metastatic phenotype in many types of tumors. We hypothesized that the αvβ3 integrin is transferred through exosomes and, upon transfer, has the ability to support functional aberrations in recipient cells. Here, for the first time, it is demonstrated that αvβ3 is present in exosomes released from metastatic PC3 and CWR22Pc prostate cancer cells. Exosomal β3 is transferred as a protein from donor to nontumorigenic and tumorigenic cells as β3 protein or mRNA levels remain unaffected upon transcription or translation inhibition in recipient cells. Furthermore, it is shown that upon exosome uptake, de novo expression of an αvβ3 increases adhesion and migration of recipient cells on an αvβ3 ligand, vitronectin. To evaluate the relevance of these findings, exosomes were purified from the blood of TRAMP mice carrying tumors where the expression of αvβ3 is found higher than in exosomes from wild-type mice. In addition, it is demonstrated that αvβ3 is coexpressed with synaptophysin, a biomarker for aggressive neuroendocrine prostate cancer. IMPLICATIONS Overall this study reveals that the αvβ3 integrin is transferred from tumorigenic to nontumorigenic cells via exosomes, and its de novo expression in recipient cells promotes cell migration on its ligand. The increased expression of αvβ3 in exosomes from mice bearing tumors points to its clinical relevance and potential use as a biomarker. Mol Cancer Res; 14(11); 1136-46. ©2016 AACR.
Collapse
Affiliation(s)
- Amrita Singh
- Prostate Cancer Discovery and Development Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Carmine Fedele
- Prostate Cancer Discovery and Development Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Huimin Lu
- Prostate Cancer Discovery and Development Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Marja T Nevalainen
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - James H Keen
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania. .,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
43
|
Simonsen TG, Gaustad JV, Rofstad EK. Intertumor heterogeneity in vascularity and invasiveness of artificial melanoma brain metastases. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:150. [PMID: 26667022 PMCID: PMC4678615 DOI: 10.1186/s13046-015-0264-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 12/01/2015] [Indexed: 12/24/2022]
Abstract
Background Patients diagnosed with melanoma brain metastases have few treatment options and poor prognosis, and improved treatment strategies for these patients require detailed understanding of the underlying pathobiology. In this investigation we studied the vascularity and invasiveness of artificial brain metastases established from four human melanoma cell lines. Methods A-07, D-12, R-18, and U-25 cells transfected with GFP were injected intracerebrally and intra-arterially in nude mice. Moribund mice were killed and autopsied, and the brain was evaluated by fluorescence imaging or by histological examination. Expression and secretion of factors involved in angiogenesis and invasion were assessed by quantitative PCR, ELISA, and immunohistochemistry. Results The melanoma cells grew preferentially in the meninges and ventricles after intracerebral and intra-arterial injection. Intertumor heterogeneity in the aggressiveness of meningeal tumors reflected differences in angiogenic activity and expression of vascular endothelial growth factor A (VEGF-A) and interleukin 8 (IL-8). In contrast, growth and invasion of the brain parenchyma relied primarily on vascular co-option. The cell lines showed different patterns of invasion from meninges to the scull and from meninges to the brain parenchyma, and these differences were associated with differences in expression of the matrix metalloproteinases MMP-2 and MMP-9. Furthermore, the melanoma cells produced multiple brain lesions after intracerebral implantation by using the meningeal linings of the brain as transport routes. Conclusions The melanoma cell lines showed different growth patterns in the brain, and these differences were associated with differences in expression of the angiogenic factors VEGF-A and IL-8 and the matrix metalloproteinases MMP-2 and MMP-9.
Collapse
Affiliation(s)
- Trude G Simonsen
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| | - Jon-Vidar Gaustad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| | - Einar K Rofstad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
44
|
Cantor DI, Cheruku HR, Nice EC, Baker MS. Integrin αvβ6 sets the stage for colorectal cancer metastasis. Cancer Metastasis Rev 2015; 34:715-34. [DOI: 10.1007/s10555-015-9591-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
45
|
Zhang X, Wu H, Dobson JR, Browne G, Hong D, Akech J, Languino LR, Stein JL, Stein GS, Lian JB. Expression of the IL-11 Gene in Metastatic Cells Is Supported by Runx2-Smad and Runx2-cJun Complexes Induced by TGFβ1. J Cell Biochem 2015; 116:2098-108. [PMID: 25808168 PMCID: PMC4515199 DOI: 10.1002/jcb.25167] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 03/18/2015] [Indexed: 12/22/2022]
Abstract
In tumor cells, two factors are abnormally increased that contribute to metastatic bone disease: Runx2, a transcription factor that promotes expression of metastasis related and osteolytic genes; and IL-11, a secreted osteolytic cytokine. Here, we addressed a compelling question: Does Runx2 regulate IL-11 gene expression? We find a positive correlation between Runx2, IL-11 and TGFβ1, a driver of the vicious cycle of metastatic bone disease, in prostate cancer (PC) cell lines representing early (LNCaP) and late (PC3) stage disease. Further, like Runx2 knockdown, IL-11 knockdown significantly reduced expression of several osteolytic factors. Modulation of Runx2 expression results in corresponding changes in IL-11 expression. The IL-11 gene has Runx2, AP-1 sites and Smad binding elements located on the IL-11 promoter. Here, we demonstrated that Runx2-c-Jun as well as Runx2-Smad complexes upregulate IL-11 expression. Functional studies identified a significant loss of IL-11 expression in PC3 cells in the presence of the Runx2-HTY mutant protein, a mutation that disrupts Runx2-Smad signaling. In response to TGFβ1 and in the presence of Runx2, we observed a 30-fold induction of IL-11 expression, accompanied by increased c-Jun binding to the IL-11 promoter. Immunoprecipitation and in situ co-localization studies demonstrated that Runx2 and c-Jun form nuclear complexes in PC3 cells. Thus, TGFβ1 signaling induces two independent transcriptional pathways - AP-1 and Runx2. These transcriptional activators converge on IL-11 as a result of Runx2-Smad and Runx2-c-Jun interactions to amplify IL-11 gene expression that, together with Runx2, supports the osteolytic pathology of cancer induced bone disease.
Collapse
Affiliation(s)
- Xuhui Zhang
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine Burlington, VT, USA
- Institute of Basic Medical Sciences, Beijing 100850, China
| | - Hai Wu
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine Burlington, VT, USA
| | - Jason R. Dobson
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Gillian Browne
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine Burlington, VT, USA
| | - Deli Hong
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine Burlington, VT, USA
| | - Jacqueline Akech
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Lucia R. Languino
- Prostate Cancer Discovery and Development Program and Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Janet L. Stein
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine Burlington, VT, USA
| | - Gary S. Stein
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine Burlington, VT, USA
| | - Jane B. Lian
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine Burlington, VT, USA
| |
Collapse
|
46
|
Wang T, Han S, Wu Z, Han Z, Yan W, Liu T, Wei H, Song D, Zhou W, Yang X, Xiao J. XCR1 promotes cell growth and migration and is correlated with bone metastasis in non-small cell lung cancer. Biochem Biophys Res Commun 2015; 464:635-641. [PMID: 26166822 DOI: 10.1016/j.bbrc.2015.06.175] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 06/23/2015] [Indexed: 01/05/2023]
Abstract
Bone metastasis occurs in approximately 30-40% patients with advanced non-small cell lung cancer (NSCLC), but the mechanism underlying this bone metastasis remains poorly understood. The chemokine super family is believed to play an important role in tumor metastasis in lung cancer. The chemokine receptor XCR1 has been identified to promote cell proliferation and migration in oral cancer and ovarian carcinoma, but the role of XCR1 in lung cancer has not been reported. In this study, we demonstrated for the first time that XCR1 was overexpressed in lung cancer bone metastasis as compared with that in patients with primary lung cancer. In addition, the XCR1 ligand XCL1 promoted the proliferation and migration of lung cancer cells markedly, and knockdown of XCR1 by siRNA abolished the effect of XCL1 in cell proliferation and migration. Furthermore, we identified JAK2/STAT3 as a novel downstream pathway of XCR1, while XCL1/XCR1 increased the mRNA level of the downstream of JAK2/STAT3 including PIM1, JunB, TTP, MMP2 and MMP9. These results indicate that XCR1 is a new potential therapeutic target for the treatment of lung cancer bone metastasis.
Collapse
Affiliation(s)
- Ting Wang
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Shuai Han
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhipeng Wu
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhitao Han
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Wangjun Yan
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Tielong Liu
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Haifeng Wei
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Dianwen Song
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Wang Zhou
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China.
| | - Xinghai Yang
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China.
| | - Jianru Xiao
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
47
|
Abstract
Transforming growth factor (TGF) β1 activity depends on a complex signalling cascade that controls expression of several genes. Among others, TGFβ1 regulates expression of matrix metalloproteinases (MMPs) through activation of Smads. In the present study, we demonstrate for the first time that the αvβ6 integrin interacts with TGFβ receptor II (TβRII) through the β6 cytoplasmic domain and promotes Smad3 activation in prostate cancer (PrCa) cells. Another related αv integrin, αvβ5, as well as the αvβ6/3 integrin, which contains a chimeric form of β6 with a β3 cytoplasmic domain, do not associate with TβRII and fail to show similar responses. We provide evidence that αvβ6 is required for up-regulation of MMP2 by TGFβ1 through a Smad3-mediated transcriptional programme in PrCa cells. The functional relevance of these results is underscored by the finding that αvβ6 modulates cell migration in an MMP2-dependent manner on an αvβ6-specific ligand, latency-associated peptide (LAP)-TGFβ. Overall, these mechanistic studies establish that expression of a single integrin, αvβ6, is sufficient to promote activation of Smad3, regulation of MMP2 levels and consequent catalytic activity, as well as cell migration. Our study describes a new TGFβ1-αvβ6-MMP2 signalling pathway that, given TGFβ1 pro-metastatic activity, may have profound implications for PrCa therapy.
Collapse
|
48
|
Wang N, Docherty F, Brown HK, Reeves K, Fowles A, Lawson M, Ottewell PD, Holen I, Croucher PI, Eaton CL. Mitotic quiescence, but not unique "stemness," marks the phenotype of bone metastasis-initiating cells in prostate cancer. FASEB J 2015; 29:3141-50. [PMID: 25888599 DOI: 10.1096/fj.14-266379] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 03/31/2015] [Indexed: 01/09/2023]
Abstract
This study aimed to identify subpopulations of prostate cancer cells that are responsible for the initiation of bone metastases. Using rapidly dividing human prostate cancer cell lines, we identified mitotically quiescent subpopulations (<1%), which we compared with the rapidly dividing populations for patterns of gene expression and for their ability to migrate to the skeletons of athymic mice. The study used 2-photon microscopy to map the presence/distribution of fluorescently labeled, quiescent cells and luciferase expression to determine the presence of growing bone metastases. We showed that the mitotically quiescent cells were very significantly more tumorigenic in forming bone metastases than fast-growing cells (55 vs. 15%) and had a unique gene expression profile. The quiescent cells were not uniquely stem cell like, with no expression of CD133 but had the same level expression of other putative prostate stem cell markers (CD44 and integrins α2/β1), when compared to the rapidly proliferating population. In addition, mitotic quiescence was associated with very high levels of C-X-C chemokine receptor type 4 (CXCR4) production. Inhibition of CXCR4 activity altered the homing of quiescent tumor cells to bone. Our studies suggest that mitotic dormancy is a unique phenotype that facilitates tumor cell colonization of the skeleton in prostate cancer.
Collapse
Affiliation(s)
- Ning Wang
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Freyja Docherty
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Hannah K Brown
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Kim Reeves
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Anne Fowles
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Michelle Lawson
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Penelope D Ottewell
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Ingunn Holen
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Peter I Croucher
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Colby L Eaton
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| |
Collapse
|
49
|
Sottnik JL, Dai J, Zhang H, Campbell B, Keller ET. Tumor-induced pressure in the bone microenvironment causes osteocytes to promote the growth of prostate cancer bone metastases. Cancer Res 2015; 75:2151-8. [PMID: 25855383 DOI: 10.1158/0008-5472.can-14-2493] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 03/25/2015] [Indexed: 01/09/2023]
Abstract
Cross-talk between tumor cells and their microenvironment is critical for malignant progression. Cross-talk mediators, including soluble factors and direct cell contact, have been identified, but roles for the interaction of physical forces between tumor cells and the bone microenvironment have not been described. Here, we report preclinical evidence that tumor-generated pressure acts to modify the bone microenvironment to promote the growth of prostate cancer bone metastases. Tumors growing in mouse tibiae increased intraosseous pressure. Application of pressure to osteocytes, the main mechanotransducing cells in bone, induced prostate cancer growth and invasion. Mechanistic investigations revealed that this process was mediated in part by upregulation of CCL5 and matrix metalloproteinases in osteocytes. Our results defined the critical contribution of physical forces to tumor cell growth in the tumor microenvironment, and they identified osteocytes as a critical mediator in the bone metastatic niche.
Collapse
Affiliation(s)
- Joseph L Sottnik
- Department of Urology, University of Michigan, Ann Arbor, Michigan.
| | - Jinlu Dai
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Honglai Zhang
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | | | - Evan T Keller
- Department of Urology, University of Michigan, Ann Arbor, Michigan. Department of Pathology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
50
|
Fedele C, Singh A, Zerlanko BJ, Iozzo RV, Languino LR. The αvβ6 integrin is transferred intercellularly via exosomes. J Biol Chem 2015; 290:4545-4551. [PMID: 25568317 DOI: 10.1074/jbc.c114.617662] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Exosomes, cell-derived vesicles of endosomal origin, are continuously released in the extracellular environment and play a key role in intercellular crosstalk. In this study, we have investigated whether transfer of integrins through exosomes between prostate cancer (PrCa) cells occurs and whether transferred integrins promote cell adhesion and migration. Among others, we have focused on the αvβ6 integrin, which is not detectable in normal human prostate but is highly expressed in human primary PrCa as well as murine PrCa in Pten(pc-/-) mice. After confirming the fidelity of the exosome preparations by electron microscopy, density gradient, and immunoblotting, we determined that the αvβ6 integrin is actively packaged into exosomes isolated from PC3 and RWPE PrCa cell lines. We also demonstrate that αvβ6 is efficiently transferred via exosomes from a donor cell to an αvβ6-negative recipient cell and localizes to the cell surface. De novo αvβ6 expression in an αvβ6-negative recipient cell is not a result of a change in mRNA levels but is a consequence of exosome-mediated transfer of this integrin between different PrCa cells. Recipient cells incubated with exosomes containing αvβ6 migrate on an αvβ6 specific substrate, latency-associated peptide-TGFβ, to a greater extent than cells treated with exosomes in which αvβ6 is stably or transiently down-regulated by shRNA or siRNA, respectively. Overall, this study shows that exosomes from PrCa cells may contribute to a horizontal propagation of integrin-associated phenotypes, which would promote cell migration, and consequently, metastasis in a paracrine fashion.
Collapse
Affiliation(s)
- Carmine Fedele
- From the Prostate Cancer Discovery and Development Program,; Department of Cancer Biology, Sidney Kimmel Cancer Center, and
| | - Amrita Singh
- From the Prostate Cancer Discovery and Development Program,; Department of Cancer Biology, Sidney Kimmel Cancer Center, and
| | - Brad J Zerlanko
- From the Prostate Cancer Discovery and Development Program,; Department of Cancer Biology, Sidney Kimmel Cancer Center, and
| | - Renato V Iozzo
- Department of Cancer Biology, Sidney Kimmel Cancer Center, and; Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107.
| | - Lucia R Languino
- From the Prostate Cancer Discovery and Development Program,; Department of Cancer Biology, Sidney Kimmel Cancer Center, and
| |
Collapse
|