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
|
Khademi R, Malekzadeh H, Bahrami S, Saki N, Khademi R, Villa-Diaz LG. Regulation and Functions of α6-Integrin (CD49f) in Cancer Biology. Cancers (Basel) 2023; 15:3466. [PMID: 37444576 DOI: 10.3390/cancers15133466] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Over the past decades, our knowledge of integrins has evolved from being understood as simple cell surface adhesion molecules to receptors that have a complex range of intracellular and extracellular functions, such as delivering chemical and mechanical signals to cells. Consequently, they actively control cellular proliferation, differentiation, and apoptosis. Dysregulation of integrin signaling is a major factor in the development and progression of many tumors. Many reviews have covered the broader integrin family in molecular and cellular studies and its roles in diseases. Nevertheless, further understanding of the mechanisms specific to an individual subunit of different heterodimers is more useful. Thus, we describe the current understanding of and exploratory investigations on the α6-integrin subunit (CD49f, VLA6; encoded by the gene itga6) in normal and cancer cells. The roles of ITGA6 in cell adhesion, stemness, metastasis, angiogenesis, and drug resistance, and as a diagnosis biomarker, are discussed. The role of ITGA6 differs based on several features, such as cell background, cancer type, and post-transcriptional alterations. In addition, exosomal ITGA6 also implies metastatic organotropism. The importance of ITGA6 in the progression of a number of cancers, including hematological malignancies, suggests its potential usage as a novel prognostic or diagnostic marker and useful therapeutic target for better clinical outcomes.
Collapse
Affiliation(s)
- Rahele Khademi
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (Immuno_TACT), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
| | - Hossein Malekzadeh
- Department of Oral Medicine, Faculty of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135715794, Iran
| | - Sara Bahrami
- Resident of Restorative Dentistry, Qazvin University of Medical Sciences, Qazvin 3419759811, Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135715794, Iran
| | - Reyhane Khademi
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (Immuno_TACT), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135715794, Iran
- Department of Medical Laboratory Sciences, School of Para-Medicine, Ahvaz Jundishapour University of Medical Sciences, Ahvaz 6135715794, Iran
| | - Luis G Villa-Diaz
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA
- Department of Bioengineering, Oakland University, Rochester, MI 48309, USA
| |
Collapse
|
3
|
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: 5.0] [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
|
4
|
Koivusalo S, Schmidt A, Manninen A, Wenta T. Regulation of Kinase Signaling Pathways by α6β4-Integrins and Plectin in Prostate Cancer. Cancers (Basel) 2022; 15:149. [PMID: 36612146 PMCID: PMC9818203 DOI: 10.3390/cancers15010149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/19/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022] Open
Abstract
Hemidesmosomes (HDs) are adhesive structures that ensure stable anchorage of cells to the basement membrane. They are formed by α6β4-integrin heterodimers and linked to intermediate filaments via plectin. It has been reported that one of the most common events during the pathogenesis of prostate cancer (PCa) is the loss of HD organization. While the expression levels of β4-integrins are strongly reduced, the expression levels of α6-integrins and plectin are maintained or even elevated, and seem to promote tumorigenic properties of PCa cells, such as proliferation, invasion, metastasis, apoptosis- and drug-resistance. In this review, we discuss the potential mechanisms of how HD components might contribute to various cellular signaling pathways to promote prostate carcinogenesis. Moreover, we summarize the current knowledge on the involvement of α6β4-integrins and plectin in PCa initiation and progression.
Collapse
Affiliation(s)
- Saara Koivusalo
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, 90220 Oulu, Finland
| | - Anette Schmidt
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, 90220 Oulu, Finland
| | - Aki Manninen
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, 90220 Oulu, Finland
| | - Tomasz Wenta
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, 90220 Oulu, Finland
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland
| |
Collapse
|
5
|
Zheng G, Bouamar H, Cserhati M, Zeballos CR, Mehta I, Zare H, Broome L, Hu R, Lai Z, Chen Y, Sharkey FE, Rani M, Halff GA, Cigarroa FG, Sun LZ. Integrin alpha 6 is upregulated and drives hepatocellular carcinoma progression through integrin α6β4 complex. Int J Cancer 2022; 151:930-943. [PMID: 35657344 PMCID: PMC9329238 DOI: 10.1002/ijc.34146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/24/2022]
Abstract
Integrin α6 (ITGA6) forms integrin receptors with either integrin β1 (ITGB1) or integrin β4 (ITGB4). How it functions to regulate hepatocellular carcinoma (HCC) progression is not well-elucidated. We found that ITGA6 RNA and protein expression levels are significantly elevated in human HCC tissues in comparison with paired adjacent nontumor tissues by RNA sequencing, RT-qPCR, Western blotting and immunofluorescence staining. Stable knockdown of ITGA6 with different ITGA6 shRNA expression lentivectors significantly inhibited proliferation, migration and anchorage-independent growth of HCC cell lines in vitro, and xenograft tumor growth in vivo. The inhibition of anchorage-dependent and -independent growth of HCC cell lines was also confirmed with anti-ITGA6 antibody. ITGA6 knockdown was shown to induce cell-cycle arrest at G0/G1 phase. Immunoprecipitation assay revealed apparent interaction of ITGA6 with ITGB4, but not ITGB1. Expression studies showed that ITGA6 positively regulates the expression of ITGB4 with no or negative regulation of ITGB1 expression. Finally, while high levels of ITGA6 and ITGB4 together were associated with significantly worse survival of HCC patients in TCGA data set, the association was not significant for high levels of ITGA6 and ITGB1. In conclusion, ITGA6 is upregulated in HCC tumors and has a malignant promoting role in HCC cells through integrin α6β4 complex. Thus, integrin α6β4 may be a therapeutic target for treating patients with HCC.
Collapse
Affiliation(s)
- Guixi Zheng
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, China
| | - Hakim Bouamar
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Matyas Cserhati
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Carla R. Zeballos
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Isha Mehta
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Habil Zare
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Larry Broome
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Ruolei Hu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Zhao Lai
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, TX
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, TX
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, TX
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, TX
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, TX
| | - Francis E. Sharkey
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, TX
| | - Meenakshi Rani
- Transplant Center, University of Texas Health Science Center at San Antonio, TX
| | - Glenn A. Halff
- Transplant Center, University of Texas Health Science Center at San Antonio, TX
| | | | - Lu-Zhe Sun
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| |
Collapse
|
6
|
Mezawa M, Tsuruya Y, Yamaguchi A, Yamazaki-Takai M, Kono T, Okada H, McCulloch CA, Ogata Y. TNF-α regulates the composition of the basal lamina and cell-matrix adhesions in gingival epithelial cells. Cell Adh Migr 2022; 16:13-24. [PMID: 35137648 PMCID: PMC8837257 DOI: 10.1080/19336918.2022.2029237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Laminin 5, type 4 collagen, and α6β4 integrin contribute to the formation of hemidesmosomes in the epithelia of periodontal tissues, which is critical for the development and maintenance of the dentogingival junction. As it is not known whether TNF-α alters the composition of the epithelial pericellular matrix, human gingival epithelial cells were cultured in the presence or absence of TNF-α. Treatment with TNF-α accelerated epithelial cell migration and closure of in vitro wounds. These data indicate unexpectedly, that TNF-α promotes the formation of the pericellular matrix around epithelial cells and enhances adhesion of epithelial cells to the underlying matrix, properties which are important for cell migration and the integrity of the dentogingival junction.
Collapse
Affiliation(s)
- Masaru Mezawa
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan.,Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Japan
| | - Yuto Tsuruya
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Arisa Yamaguchi
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Mizuho Yamazaki-Takai
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Tetsuro Kono
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Japan.,Department of Histology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Hiroyuki Okada
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Japan.,Department of Histology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | | | - Yorimasa Ogata
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan.,Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Japan
| |
Collapse
|
7
|
Integrin expression in correlation to clinicopathological features and prognosis of prostate cancer: A systematic review and meta-analysis. Urol Oncol 2021; 39:221-232. [PMID: 33558138 DOI: 10.1016/j.urolonc.2020.12.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/20/2020] [Accepted: 12/22/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND The prompt identification of patients with poor prognosis is essential in order to improve the treatment outcomes in prostate cancer (CaP); as a novel approach, several molecular markers, including integrins, have been discussed as prognostic biomarkers. Our aim was to comprehensively examine aberrant expression of integrins in correlation with clinicopathological features and prognosis in CaP by synthesizing all available evidence, in a systematic review and meta-analysis. METHODS A systematic review and meta-analysis was performed in accordance with the Preferred Reporting Items for Systematic Review and Meta-analysis (PRISMA) guidelines. Scientific literature databases (Pubmed, Embase, and Scopus) were systematically searched until May 10, 2020. Random-effects (DerSimonian-Laird) models were used to estimate pooled odds ratios (ORs) for cross-sectional correlations with clinicopathological characteristics and relative risks for longitudinal associations with prognosis. RESULTS Fourteen studies were included with a total number of 3,194 CaP cases examined (13 cross-sectional and four longitudinal cohort study arms). Correlation of low expression of α6 (pooled OR = 0.10, 95% confidence interval [CI]: 0.04-0.28, P < 0.001) and β1 (pooled OR = 0.45; 95% CI: 0.21-1.00, P = 0.049) integrin with high Gleason score was noted. A borderline trend between reduced expression of α6 integrin and an advanced clinical stage of CaP (pooled OR = 0.48; 95% CI: 0.22-1.03, P = 0.06) was observed. No associations with biochemical recurrence and survival were documented. CONCLUSIONS Evidence on the association of low expression of integrins α6 and β1 and more advanced CaP exist, whereas significant results on survival were not documented; further studies are warranted.
Collapse
|
8
|
Cohesive cancer invasion of the biophysical barrier of smooth muscle. Cancer Metastasis Rev 2021; 40:205-219. [PMID: 33398621 DOI: 10.1007/s10555-020-09950-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 12/15/2020] [Indexed: 01/22/2023]
Abstract
Smooth muscle is found around organs in the digestive, respiratory, and reproductive tracts. Cancers arising in the bladder, prostate, stomach, colon, and other sites progress from low-risk disease to high-risk, lethal metastatic disease characterized by tumor invasion into, within, and through the biophysical barrier of smooth muscle. We consider here the unique biophysical properties of smooth muscle and how cohesive clusters of tumor use mechanosensing cell-cell and cell-ECM (extracellular matrix) adhesion receptors to move through a structured muscle and withstand the biophysical forces to reach distant sites. Understanding integrated mechanosensing features within tumor cluster and smooth muscle and potential triggers within adjacent adipose tissue, such as the unique damage-associated molecular pattern protein (DAMP), eNAMPT (extracellular nicotinamide phosphoribosyltransferase), or visfatin, offers an opportunity to prevent the first steps of invasion and metastasis through the structured muscle.
Collapse
|
9
|
Jales Neto LH, Wicik Z, Torres GHF, Takayama L, Caparbo VF, Lopes NHM, Pereira AC, Pereira RMR. Overexpression of SNTG2, TRAF3IP2, and ITGA6 transcripts is associated with osteoporotic vertebral fracture in elderly women from community. Mol Genet Genomic Med 2020; 8:e1391. [PMID: 32602654 PMCID: PMC7507059 DOI: 10.1002/mgg3.1391] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/28/2020] [Accepted: 06/01/2020] [Indexed: 12/19/2022] Open
Abstract
Background Vertebral fractures (VFs) are the most common clinical manifestation of osteoporosis associated with high morbimortality. A personal/familiar history of fractures increases the risk of fractures. The purpose of this study is to identify possible molecular markers associated with osteoporotic VFs in elderly women from community. Methods Transcriptomic analysis using Affymetrix HTA2 microarray was performed using whole blood samples of 240 subjects from a population‐based survey (Sao Paulo Ageing & Health [SPAH] study). Only elderly women with osteoporosis diagnosis by densitometry were analyzed, and divided in two groups: VF: women with osteoporosis and VFs versus no vertebral fracture (NVF): women with osteoporosis and NVFs. They were matched for age, chronic disease, medication use, and bone mineral density (BMD). The logistic regression model adjusted for age was applied for transcriptome data analysis. SYBR green‐based quantitative polymerase chain reaction (qPCR) was used to validate the most significant expression changes obtained in the microarray experiment. Results Microarray analysis identified 142 differentially expressed genes (DEGs, p < .01), 57 upregulated and 85 downregulated, compared VF versus NVF groups. The DEG with the greatest expression difference was the Gamma2‐Syntrophin (SNTG2) (β = 31.88, p = .005). Validation by qPCR confirmed increased expression in VF group of Syntrophin (SNTG2, fold change = 2.79, p = .009), TRAF3 Interacting Protein2 (TRAF3IP2, fold change = 2.79, p = .020), and Integrin Subunit Alpha 6 (ITGA6, fold change = 2.86, p = .038). Conclusion Our data identified and validated the association of SNTG2 (608715), TRAF3IP2 (607043), and ITGA6 (147556) with osteoporotic VF in elderly women, independently of BMD. These results suggest that these transcripts have potential clinical significance and may help to explain the molecular mechanisms and biological functions of vertebral fracture.
Collapse
Affiliation(s)
- Levi H Jales Neto
- Bone Metabolism Laboratory, Rheumatology Division Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Zofia Wicik
- Bone Metabolism Laboratory, Rheumatology Division Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Georgea H F Torres
- Bone Metabolism Laboratory, Rheumatology Division Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Liliam Takayama
- Bone Metabolism Laboratory, Rheumatology Division Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Valéria F Caparbo
- Bone Metabolism Laboratory, Rheumatology Division Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Neuza H M Lopes
- Faculdade de Medicina, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology, Faculdade de Medicina, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Rosa M R Pereira
- Bone Metabolism Laboratory, Rheumatology Division Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
10
|
MicroRNA-377-3p alleviates IL-1β-caused chondrocyte apoptosis and cartilage degradation in osteoarthritis in part by downregulating ITGA6. Biochem Biophys Res Commun 2019; 523:46-53. [PMID: 31831175 DOI: 10.1016/j.bbrc.2019.11.186] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 11/28/2019] [Indexed: 12/23/2022]
Abstract
Increasing evidence indicates that altered expression of microRNAs (miRNAs) is associated with osteoarthritis (OA) progression. In our study, we demonstrated that miR-377-3p is underexpressed in OA-affected cartilage and IL-1β-treated chondrocytes. Overexpression of miR-377-3p enhanced chondrocyte proliferation and restrained apoptosis and signs of cartilage matrix degradation and of an inflammatory response. Furthermore, ITGA6 was identified as a target gene of miR-377-3p. The latter was found to directly bind to the 3' untranslated region (3'UTR) of ITGA6 mRNA and downregulate ITGA6. In addition, ITGA6 expression was high in OA-affected tissues and negatively correlated with miR-77-3p expression. Overexpression of ITGA6 reversed the effects of miR-377-3p on IL-1β-caused chondrocyte apoptosis, cartilage matrix degradation, and the inflammatory response. Moreover, bioinformatic analysis and a luciferase assay indicated that miR-377-3p expression is regulated by long noncoding RNA NEAT1, which binds to miR-377-3p and inactivates it. We showed that NEAT1 was highly expressed in OA-affected cartilage, negatively correlated with miR-377-3p levels, and positively correlated with ITGA6 levels. These findings provide information for the development of future treatments of OA, suggesting that miR-377-3p may be a therapeutic target in OA.
Collapse
|
11
|
Hinton JP, Dvorak K, Roberts E, French WJ, Grubbs JC, Cress AE, Tiwari HA, Nagle RB. A Method to Reuse Archived H&E Stained Histology Slides for a Multiplex Protein Biomarker Analysis. Methods Protoc 2019; 2:mps2040086. [PMID: 31731599 PMCID: PMC6960855 DOI: 10.3390/mps2040086] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022] Open
Abstract
Archived Hematoxylin and Eosin (H&E) stained pathology slides are routinely stored to index formalin-fixed paraffin-embedded (FFPE) sample tissue blocks. FFPE blocks are clinically annotated human tumor specimens that can be valuable in studies decades after the tissue is collected. If stored properly, they have the potential to yield a valuable number of serial sectioned slides for diagnostic or research purposes. However, some retrospective studies are limited in scope because the tissue samples have been depleted or not enough material is available in stored blocks for serial sections. The goal of these studies was to determine if archived H&E-stained slides can be directly reutilized by optimizing methods to de-stain and then re-stain the H&E stained slides to allow the detection of several biomarkers of interest using a conjugated antibody with chromogen multiplex immunohistochemistry procedure. This simple but innovative procedure, combined with image analysis techniques, demonstrates the ability to perform precise detection of relevant markers correlated to disease progression in initially identified tumor regions in tissue. This may add clinical value in retaining H&E slides for further use.
Collapse
Affiliation(s)
- James P. Hinton
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona Cancer Center, Tucson, AZ 85724, USA;
- Ventana/Roche Tissue Diagnostics, Tucson, AZ 85755, USA; (K.D.); (E.R.); (W.J.F.); (J.C.G.)
| | - Katerina Dvorak
- Ventana/Roche Tissue Diagnostics, Tucson, AZ 85755, USA; (K.D.); (E.R.); (W.J.F.); (J.C.G.)
| | - Esteban Roberts
- Ventana/Roche Tissue Diagnostics, Tucson, AZ 85755, USA; (K.D.); (E.R.); (W.J.F.); (J.C.G.)
| | - Wendy J. French
- Ventana/Roche Tissue Diagnostics, Tucson, AZ 85755, USA; (K.D.); (E.R.); (W.J.F.); (J.C.G.)
| | - Jon C. Grubbs
- Ventana/Roche Tissue Diagnostics, Tucson, AZ 85755, USA; (K.D.); (E.R.); (W.J.F.); (J.C.G.)
| | - Anne E. Cress
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona Cancer Center, Tucson, AZ 85724, USA;
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
- Correspondence: ; Tel.: +1-520-626-7553
| | - Hina A. Tiwari
- Department of Medical Imaging, College of Medicine, University of Arizona, Tucson, AZ 85724, USA;
| | - Raymond B. Nagle
- Department of Pathology, College of Medicine, the University of Arizona, Tucson, AZ 85724, USA;
| |
Collapse
|
12
|
Rubenstein CS, Gard JMC, Wang M, McGrath JE, Ingabire N, Hinton JP, Marr KD, Simpson SJ, Nagle RB, Miranti CK, Warfel NA, Garcia JGN, Arif-Tiwari H, Cress AE. Gene Editing of α6 Integrin Inhibits Muscle Invasive Networks and Increases Cell-Cell Biophysical Properties in Prostate Cancer. Cancer Res 2019; 79:4703-4714. [PMID: 31337652 PMCID: PMC6750953 DOI: 10.1158/0008-5472.can-19-0868] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 06/10/2019] [Accepted: 07/19/2019] [Indexed: 12/26/2022]
Abstract
Human prostate cancer confined to the gland is indolent (low-risk), but tumors outside the capsule are aggressive (high-risk). Extracapsular extension requires invasion within and through a smooth muscle-structured environment. Because integrins respond to biomechanical cues, we used a gene editing approach to determine if a specific region of laminin-binding α6β1 integrin was required for smooth muscle invasion both in vitro and in vivo. Human tissue specimens showed prostate cancer invasion through smooth muscle and tumor coexpression of α6 integrin and E-cadherin in a cell-cell location and α6 integrin in a cell-extracellular matrix (ECM) distribution. Prostate cancer cells expressing α6 integrin (DU145 α6WT) produced a 3D invasive network on laminin-containing Matrigel and invaded into smooth muscle both in vitro and in vivo. In contrast, cells without α6 integrin (DU145 α6KO) and cells expressing an integrin mutant (DU145 α6AA) did not produce invasive networks, could not invade muscle both in vitro and in vivo, and surprisingly formed 3D cohesive clusters. Using electric cell-substrate impedance testing, cohesive clusters had up to a 30-fold increase in normalized resistance at 400 Hz (cell-cell impedance) as compared with the DU145 α6WT cells. In contrast, measurements at 40,000 Hz (cell-ECM coverage) showed that DU145 α6AA cells were two-fold decreased in normalized resistance and were defective in restoring resistance after a 1 μmol/L S1P challenge as compared with the DU145 α6WT cells. The results suggest that gene editing of a specific α6 integrin extracellular region, not required for normal tissue function, can generate a new biophysical cancer phenotype unable to invade the muscle, presenting a new therapeutic strategy for metastasis prevention in prostate cancer. SIGNIFICANCE: This study shows an innovative strategy to block prostate cancer metastasis and invasion in the muscle through gene editing of a specific α6 integrin extracellular region.
Collapse
Affiliation(s)
| | - Jaime M C Gard
- Cancer Biology Research Program, University of Arizona, Tucson, Arizona
| | - Mengdie Wang
- Cancer Biology Research Program, University of Arizona, Tucson, Arizona
| | - Julie E McGrath
- Cancer Biology Research Program, University of Arizona, Tucson, Arizona
| | - Nadia Ingabire
- Cancer Biology Research Program, University of Arizona, Tucson, Arizona
| | - James P Hinton
- Cancer Biology Research Program, University of Arizona, Tucson, Arizona
| | - Kendra D Marr
- Cancer Biology Research Program, University of Arizona, Tucson, Arizona
| | - Skyler J Simpson
- Cancer Biology Research Program, University of Arizona, Tucson, Arizona
| | - Raymond B Nagle
- Department of Pathology, University of Arizona, Tucson, Arizona
| | - Cindy K Miranti
- Cancer Biology Research Program, University of Arizona, Tucson, Arizona
- Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Noel A Warfel
- Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Joe G N Garcia
- Department of Medicine, University of Arizona, Tucson, Arizona
| | - Hina Arif-Tiwari
- Medical Imaging and the University of Arizona Cancer Center, University of Arizona, Tucson, Arizona
| | - Anne E Cress
- Cancer Biology Research Program, University of Arizona, Tucson, Arizona.
- Department of Pathology, University of Arizona, Tucson, Arizona
- Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
- Radiation Oncology, University of Arizona, Tucson, Arizona
| |
Collapse
|
13
|
Feng GK, Ye JC, Zhang WG, Mei Y, Zhou C, Xiao YT, Li XL, Fan W, Wang F, Zeng MS. Integrin α6 targeted positron emission tomography imaging of hepatocellular carcinoma in mouse models. J Control Release 2019; 310:11-21. [PMID: 31400382 DOI: 10.1016/j.jconrel.2019.08.003] [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] [Received: 04/22/2019] [Revised: 07/13/2019] [Accepted: 08/03/2019] [Indexed: 12/20/2022]
Abstract
Integrin α6 emerges to be a diagnostic biomarker for hepatocellular carcinoma (HCC). Here, we translated our previously identified integrin α6 targeted peptide RWY into a positron emission tomography (PET) tracer 18F-RWY for the detection of HCC lesions in following four HCC mouse models including subcutaneous, orthotopic, genetically engineered and chemical induced HCC mice. 18F-RWY produced high PET signals in liver tumor tissues that were reduced by blocking studies using nonradiolabeled RWY peptide. We compared the integrin α6 targeted PET tracer 18F-RWY with the integrin αvβ3-targeted PET tracer 18F-3PRGD2 and the clinical PET tracer 18F-FDG in chemical induced HCC mice. Among 12 HCC identified by enhanced magnetic resonance imaging (MRI) with hepatocellular specific gadoxetate disodium Gd-EOB-DTPA, the sensitivities of 18F-RWY, 18F-3PRGD2 and 18F-FDG were approximately 92%, 73% and 50% while the tumor-to-liver ratios were 4.36 ± 1.41, 1.97 ± 0.43 and 1.63 ± 0.23 respectively. Additionally, PET imaging with the integrin α6 targeted 18F-RWY enabled to visualize small HCC lesions with diameters approximately 0.2 cm that was hard to be distinguished from surround hepatic vascular by enhanced MRI with Gd-EOB-DTPA. These findings potentiate the use of integrin α6 targeted PET tracer 18F-RWY for the detection of HCC.
Collapse
Affiliation(s)
- Guo-Kai Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jia-Cong Ye
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Wei-Guang Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yan Mei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chao Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yi-Tai Xiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xin-Ling Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Wei Fan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Fan Wang
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China.
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
14
|
Mani J, Neuschäfer J, Resch C, Rutz J, Maxeiner S, Roos F, Chun FKH, Juengel E, Blaheta RA. Amygdalin Modulates Prostate Cancer Cell Adhesion and Migration In Vitro. Nutr Cancer 2019; 72:528-537. [DOI: 10.1080/01635581.2019.1637442] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Jens Mani
- Department of Urology, Goethe-University, Frankfurt am Main, Germany
| | - Jens Neuschäfer
- Department of Urology, Goethe-University, Frankfurt am Main, Germany
| | - Christian Resch
- Department of Urology, Goethe-University, Frankfurt am Main, Germany
| | - Jochen Rutz
- Department of Urology, Goethe-University, Frankfurt am Main, Germany
| | | | - Frederik Roos
- Department of Urology, Goethe-University, Frankfurt am Main, Germany
| | - Felix K.-H. Chun
- Department of Urology, Goethe-University, Frankfurt am Main, Germany
| | - Eva Juengel
- Department of Urology, Goethe-University, Frankfurt am Main, Germany
| | - Roman A. Blaheta
- Department of Urology, Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|
15
|
Feng G, Zhang M, Wang H, Cai J, Chen S, Wang Q, Gong J, Leong KW, Wang J, Zhang X, Zeng M. Identification of an Integrin α6‐Targeted Peptide for Nasopharyngeal Carcinoma‐Specific Nanotherapeutics. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Guo‐Kai Feng
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center Guangzhou Guangdong 510060 P. R. China
| | - Meng‐Qing Zhang
- Rehabilitation Departmentthe Third Affiliated Hospital of Sun Yat‐sen University Guangzhou Guangdong 510060 P. R. China
| | - Hong‐Xia Wang
- Department of Biomedical EngineeringColumbia University New York NY 10027 USA
| | - Jing Cai
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center Guangzhou Guangdong 510060 P. R. China
| | - Shu‐Peng Chen
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center Guangzhou Guangdong 510060 P. R. China
| | - Qian Wang
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center Guangzhou Guangdong 510060 P. R. China
| | - Jing Gong
- Department of Biomedical EngineeringColumbia University New York NY 10027 USA
| | - Kam W. Leong
- Department of Biomedical EngineeringColumbia University New York NY 10027 USA
| | - Jun Wang
- Hefei National Laboratory for Physical Sciences at the MicroscaleUniversity of Science and Technology of China Hefei Anhui 230027 P. R. China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of Technology Guangzhou Guangdong 510006 P. R. China
| | - Xing Zhang
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center Guangzhou Guangdong 510060 P. R. China
| | - Mu‐Sheng Zeng
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center Guangzhou Guangdong 510060 P. R. China
| |
Collapse
|
16
|
Harryman WL, Warfel NA, Nagle RB, Cress AE. The Tumor Microenvironments of Lethal Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:149-170. [PMID: 31900909 DOI: 10.1007/978-3-030-32656-2_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Localized prostate cancer (confined to the gland) generally is considered curable, with nearly a 100% 5-year-survival rate. When the tumor escapes the prostate capsule, leading to metastasis, there is a poorer prognosis and higher mortality rate, with 5-year survival dropping to less than 30%. A major research question has been to understand the transition from indolent (low risk) disease to aggressive (high risk) disease. In this chapter, we provide details of the changing tumor microenvironments during prostate cancer invasion and their role in the progression and metastasis of lethal prostate cancer. Four microenvironments covered here include the muscle stroma, perineural invasion, hypoxia, and the role of microvesicles in altering the extracellular matrix environment. The adaptability of prostate cancer to these varied microenvironments and the cues for phenotypic changes are currently understudied areas. Model systems for understanding smooth muscle invasion both in vitro and in vivo are highlighted. Invasive human needle biopsy tissue and mouse xenograft tumors both contain smooth muscle invasion. In combination, the models can be used in an iterative process to validate molecular events for smooth muscle invasion in human tissue. Understanding the complex and interacting microenvironments in the prostate holds the key to early detection of high-risk disease and preventing tumor invasion through escape from the prostate capsule.
Collapse
Affiliation(s)
| | - Noel A Warfel
- University of Arizona Cancer Center, Tucson, AZ, USA
| | - Raymond B Nagle
- Department of Pathology, University of Arizona Cancer Center, Tucson, AZ, USA
| | - Anne E Cress
- University of Arizona Cancer Center, Tucson, AZ, USA.
| |
Collapse
|
17
|
Kowalski-Chauvel A, Modesto A, Gouaze-Andersson V, Baricault L, Gilhodes J, Delmas C, Lemarie A, Toulas C, Cohen-Jonathan-Moyal E, Seva C. Alpha-6 integrin promotes radioresistance of glioblastoma by modulating DNA damage response and the transcription factor Zeb1. Cell Death Dis 2018; 9:872. [PMID: 30158599 PMCID: PMC6115442 DOI: 10.1038/s41419-018-0853-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 12/30/2022]
Abstract
Radiotherapy is the cornerstone of glioblastoma (GBM) standard treatment. However, radioresistance of cancer cells leads to an inevitable recurrence. In the present study, we showed that blocking α6-integrin in cells derived from GBM biopsy specimens cultured as neurospheres, sensitized cells to radiation. In cells downregulated for α6-integrin expression, we observed a decrease in cell survival after irradiation and an increase in radio-induced cell death. We also demonstrated that inhibition of α6-integrin expression affects DNA damage checkpoint and repair. Indeed, we observed a persistence of γ-H2AX staining after IR and the abrogation of the DNA damage-induced G2/M checkpoint, likely through the downregulation of the checkpoint kinase CHK1 and its downstream target Cdc25c. We also showed that α6-integrin contributes to GBM radioresistance by controlling the expression of the transcriptional network ZEB1/OLIG2/SOX2. Finally, the clinical data from TCGA and Rembrandt databases demonstrate that GBM patients with high levels of the five genes signature, including α6-integrin and its targets, CHK1, ZEB1, OLIG2 and SOX2, have a significantly shorter overall survival. Our study suggest that α6-integrin is an attractive therapeutic target to overcome radioresistance of GBM cancer cells.
Collapse
Affiliation(s)
- Aline Kowalski-Chauvel
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier, Toulouse III, France
| | - Anouchka Modesto
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier, Toulouse III, France
- IUCT-oncopole, Toulouse, France
| | - Valerie Gouaze-Andersson
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier, Toulouse III, France
| | - Laurent Baricault
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier, Toulouse III, France
| | | | - Caroline Delmas
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier, Toulouse III, France
- IUCT-oncopole, Toulouse, France
| | - Anthony Lemarie
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier, Toulouse III, France
| | - Christine Toulas
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier, Toulouse III, France
- IUCT-oncopole, Toulouse, France
| | - Elizabeth Cohen-Jonathan-Moyal
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier, Toulouse III, France
- IUCT-oncopole, Toulouse, France
| | - Catherine Seva
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier, Toulouse III, France.
| |
Collapse
|
18
|
Xie S, Yu X, Li Y, Ma H, Fan S, Chen W, Pan G, Wang W, Zhang H, Li J, Lin Z. Upregulation of lncRNA ADAMTS9-AS2 Promotes Salivary Adenoid Cystic Carcinoma Metastasis via PI3K/Akt and MEK/Erk Signaling. Mol Ther 2018; 26:2766-2778. [PMID: 30217729 DOI: 10.1016/j.ymthe.2018.08.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 08/06/2018] [Accepted: 08/19/2018] [Indexed: 01/10/2023] Open
Abstract
Neurotropic infiltrative growth and distant metastasis are the main causes of death in salivary adenoid cystic carcinoma (SACC) patients. Long noncoding RNAs (lncRNAs) are involved in many human neoplasms, however, their potential roles in SACC are unclear. In our study, we found that ADAM metallopeptidase with thrombospondin type 1 motif, 9 (ADAMTS9) antisense RNA 2 (ADAMTS9-AS2) was significantly upregulated in SACC patients with metastasis and SACC-lung metastasis (LM) cells. Moreover, ADAMTS9-AS2 expression was closely associated with the prognosis and distant metastasis in SACC patients. Next, we found that c-myc could specifically bind to the promoter of ADAMTS9-AS2 and activated its transcription. Knockdown of ADAMTS9-AS2 significantly inhibited migration and invasion of SACC cells in vitro and distant lung metastasis in vivo. Furthermore, ADAMTS9-AS2, which mainly expressed in the cytoplasm, shared microRNA (miRNA) response elements with Integrin α6 (ITGA6). Overexpression of ADAMTS9-AS2 competitively bound to miR-143-3p that inhibited ITGA6 from miRNA-mediated degradation, and thus it activated the activity of PI3K/Akt and MEK/Erk signaling and facilitated SACC metastasis. In summary, ADAMTS9-AS2 promotes migration and invasion in SACC by competing with miR-143-3p. This sheds a new insight into the regulation mechanism of ADAMTS9-AS2, and it provides a possible application for the SACC treatment.
Collapse
Affiliation(s)
- Shule Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xin Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yingru Li
- Department of Gastroenterology, Hernia and Abdominal Wall Surgery, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hanyu Ma
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Song Fan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Weixiong Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Guokai Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Weiwei Wang
- Department of Stomatology, Zibo Center Hospital, Zibo, China
| | - Hanqing Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jinsong Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Zhaoyu Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
19
|
Ricci F, Fratelli M, Guffanti F, Porcu L, Spriano F, Dell'Anna T, Fruscio R, Damia G. Patient-derived ovarian cancer xenografts re-growing after a cisplatinum treatment are less responsive to a second drug re-challenge: a new experimental setting to study response to therapy. Oncotarget 2018; 8:7441-7451. [PMID: 26910918 PMCID: PMC5352333 DOI: 10.18632/oncotarget.7465] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/05/2016] [Indexed: 02/06/2023] Open
Abstract
Even if ovarian cancer patients are very responsive to a cisplatinum-based therapy, most will relapse with a resistant disease. New experimental animal models are needed to explore the mechanisms of resistance, to better tailor treatment and improve patient prognosis. To address these aims, seven patient-derived high-grade serous/endometrioid ovarian cancer xenografts were characterized for the antitumor response after one and two cycles of cisplatinum and classified as Very Responsive, Responsive, and Low Responsive to drug treatment. Xenografts re-growing after the first drug cycle were much less responsive to the second one. The expression of epithelial-mesenchymal transition (EMT) and cancer stem cells (CSCs) genes was investigated in cisplatinum-treated and not-treated tumors. We found that different EMT (TCF3, CAMK2N1, EGFR, and IGFBP4) and CSCs (SMO, DLL1, STAT3, and ITGA6) genes were expressed at higher levels in Low Responsive than in Responsive and Very Responsive xenografts. The expression of STAT3 was found to be associated with lower survival (HR = 13.7; p = 0.013) in the TCGA patient data set. MMP9, CD44, DLL4, FOXP1, MERTK, and PTPRC genes were found more expressed in tumors re-growing after cisplatinum treatment than in untreated tumors. We here describe a new in vivo ovarian carcinoma experimental setting that will be instrumental for specific trials of combination therapy to counteract cisplatinum resistance in order to improve the prognosis of ovarian patients.
Collapse
Affiliation(s)
- Francesca Ricci
- Department of Oncology, Laboratory of Molecular Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Maddalena Fratelli
- Department of Biochemistry, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Federica Guffanti
- Department of Oncology, Laboratory of Molecular Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Luca Porcu
- Department of Oncology, Laboratory of Methodology for Biomedical Research, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Filippo Spriano
- Department of Oncology, Laboratory of Molecular Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Tiziana Dell'Anna
- Obstetrics and Gynecology Clinic, San Gerardo Hospital, Monza, Italy
| | - Robert Fruscio
- Obstetrics and Gynecology Clinic, San Gerardo Hospital, Monza, Italy
| | - Giovanna Damia
- Department of Oncology, Laboratory of Molecular Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
20
|
Raab-Westphal S, Marshall JF, Goodman SL. Integrins as Therapeutic Targets: Successes and Cancers. Cancers (Basel) 2017; 9:E110. [PMID: 28832494 PMCID: PMC5615325 DOI: 10.3390/cancers9090110] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 12/12/2022] Open
Abstract
Integrins are transmembrane receptors that are central to the biology of many human pathologies. Classically mediating cell-extracellular matrix and cell-cell interaction, and with an emerging role as local activators of TGFβ, they influence cancer, fibrosis, thrombosis and inflammation. Their ligand binding and some regulatory sites are extracellular and sensitive to pharmacological intervention, as proven by the clinical success of seven drugs targeting them. The six drugs on the market in 2016 generated revenues of some US$3.5 billion, mainly from inhibitors of α4-series integrins. In this review we examine the current developments in integrin therapeutics, especially in cancer, and comment on the health economic implications of these developments.
Collapse
Affiliation(s)
- Sabine Raab-Westphal
- Translational In Vivo Pharmacology, Translational Innovation Platform Oncology, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany.
| | - John F Marshall
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| | - Simon L Goodman
- Translational and Biomarkers Research, Translational Innovation Platform Oncology, Merck KGaA, 64293 Darmstadt, Germany.
| |
Collapse
|
21
|
Jiang P, Li Z, Tian F, Li X, Yang J. Fyn/heterogeneous nuclear ribonucleoprotein E1 signaling regulates pancreatic cancer metastasis by affecting the alternative splicing of integrin β1. Int J Oncol 2017; 51:169-183. [PMID: 28560430 PMCID: PMC5467783 DOI: 10.3892/ijo.2017.4018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/18/2017] [Indexed: 01/29/2023] Open
Abstract
Pancreatic cancer is characterized by a dense desmoplastic reaction in which extracellular matrix proteins accumulate and surround tumor cells. Integrins and their related signaling molecules are associated with progression of pancreatic cancer. In the present study, the association between the metastasis of pancreatic cancer and the expression of hnRNP E1 and integrin β1 was evaluated. In vitro and in vivo experiments were designed to study the mechanism underlying the regulation of integrin β1 splicing by the Fyn/hnRNP E1 spliceosome. Expression of hnRNP E1 and integrin β1A were associated with metastasis of pancreatic cancer. Inhibition of Fyn activity upregulated the expression of P21-activated kinase 1 and promoted the phosphorylation and nuclear localization of hnRNP E1, leading to the construction of a spliceosome complex that affected the alterative splicing of integrin β1. In the hnRNP E1 spliceosome complex, hnRNP A1 and serine/arginine-rich splicing factor 1 were responsible for binding to the pre-mRNA of integrin β1. Suppression of Fyn activity and/or overexpression of hnRNP E1 decreased the metastasis of pancreatic cancer cells. In pancreatic cancer, the present study demonstrated a novel mechanism by which Fyn/hnRNP E1 signaling regulates pancreatic cancer metastasis by affecting the alternative splicing of integrin β1. hnRNP E1 and integrin β1A are associated with the metastasis of pancreatic cancer and may be novel molecular targets for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Peng Jiang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Zhonghu Li
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Feng Tian
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Xiaowu Li
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jin Yang
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
22
|
Das L, Anderson TA, Gard JM, Sroka IC, Strautman SR, Nagle RB, Morrissey C, Knudsen BS, Cress AE. Characterization of Laminin Binding Integrin Internalization in Prostate Cancer Cells. J Cell Biochem 2017; 118:1038-1049. [PMID: 27509031 PMCID: PMC5553695 DOI: 10.1002/jcb.25673] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 08/09/2016] [Indexed: 12/27/2022]
Abstract
Laminin binding integrins α6 (CD49f) and α3 (CD49c) are persistently but differentially expressed in prostate cancer (PCa). Integrin internalization is an important determinant of their cell surface expression and function. Using flow cytometry, and first order kinetic modeling, we quantitated the intrinsic internalization rates of integrin subunits in a single cycle of internalization. In PCa cell line DU145, α6 integrin internalized with a rate constant (kactual ) of 3.25 min-1 , threefold faster than α3 integrin (1.0 min-1 ), 1.5-fold faster than the vitronectin binding αv integrin (CD51) (2.2 min-1 ), and significantly slower than the unrelated transferrin receptor (CD71) (15 min-1 ). Silencing of α3 integrin protein expression in DU145, PC3, and PC3B1 cells resulted in up to a 1.71-fold increase in kactual for α6 integrin. The internalized α6 integrin was targeted to early endosomes but not to lamp1 vesicles. Depletion of α3 integrin expression resulted in redistribution of α6β4 integrin to an observed cell-cell staining pattern that is consistent with a suprabasal distribution observed in epidermis and early PIN lesions in PCa. Depletion of α3 integrin increased cell migration by 1.8-fold, which was dependent on α6β1 integrin. Silencing of α6 integrin expression however, had no significant effect on the kactual of α3 integrin or its distribution in early endosomes. These results indicate that α3 and α6 integrins have significantly different internalization kinetics and that coordination exists between them for internalization. J. Cell. Biochem. 118: 1038-1049, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lipsa Das
- Department of Cancer Biology, University of Arizona, Tucson, AZ 85724
| | - Todd A. Anderson
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | - Jaime M.C. Gard
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | - Isis C. Sroka
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724
| | | | - Raymond B. Nagle
- Department of Pathology, University of Arizona, Tucson, AZ 85724
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | | | | | - Anne E. Cress
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85724
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| |
Collapse
|
23
|
Harryman WL, Hinton JP, Rubenstein CP, Singh P, Nagle RB, Parker SJ, Knudsen BS, Cress AE. The Cohesive Metastasis Phenotype in Human Prostate Cancer. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1866:221-231. [PMID: 27678419 PMCID: PMC5534328 DOI: 10.1016/j.bbcan.2016.09.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 08/27/2016] [Accepted: 09/23/2016] [Indexed: 12/21/2022]
Abstract
A critical barrier for the successful prevention and treatment of recurrent prostate cancer is detection and eradication of metastatic and therapy-resistant disease. Despite the fall in diagnoses and mortality, the reported incidence of metastatic disease has increased 72% since 2004. Prostate cancer arises in cohesive groups as intraepithelial neoplasia, migrates through muscle and leaves the gland via perineural invasion for hematogenous dissemination. Current technological advances have shown cohesive-clusters of tumor (also known as microemboli) within the circulation. Circulating tumor cell (CTC) profiles are indicative of disseminated prostate cancer, and disseminated tumor cells (DTC) are found in cohesive-clusters, a phenotypic characteristic of both radiation- and drug-resistant tumors. Recent reports in cell biology and informatics, coupled with mass spectrometry, indicate that the integrin adhesome network provides an explanation for the biophysical ability of cohesive-clusters of tumor cells to invade thorough muscle and nerve microenvironments while maintaining adhesion-dependent therapeutic resistance. Targeting cohesive-clusters takes advantage of the known ability of extracellular matrix (ECM) adhesion to promote tumor cell survival and represents an approach that has the potential to avoid the progression to drug- and radiotherapy-resistance. In the following review we will examine the evidence for development and dissemination of cohesive-clusters in metastatic prostate cancer.
Collapse
Affiliation(s)
- William L Harryman
- The University of Arizona Cancer Center, 1515 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - James P Hinton
- Cancer Biology Graduate Program, The University of Arizona Cancer Center, 1515 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Cynthia P Rubenstein
- Cancer Biology Graduate Program, The University of Arizona Cancer Center, 1515 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Parminder Singh
- The University of Arizona Cancer Center, 1515 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Raymond B Nagle
- The University of Arizona Cancer Center, 1515 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Sarah J Parker
- Cedars Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Beatrice S Knudsen
- Cedars Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Anne E Cress
- The University of Arizona Cancer Center, 1515 N. Campbell Ave., Tucson, AZ, 85724, USA.
| |
Collapse
|
24
|
Sroka IC, Chopra H, Das L, Gard JMC, Nagle RB, Cress AE. Schwann Cells Increase Prostate and Pancreatic Tumor Cell Invasion Using Laminin Binding A6 Integrin. J Cell Biochem 2016; 117:491-9. [PMID: 26239765 DOI: 10.1002/jcb.25300] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 07/31/2015] [Indexed: 01/13/2023]
Abstract
Human pancreatic and prostate cancers metastasize along nerve axons during perineural invasion. The extracellular matrix laminin class of proteins is an abundant component of both myelinated and non-myelinated nerves. Analysis of human pancreatic and prostate tissue revealed both perineural and endoneural invasion with Schwann cells surrounded or disrupted by tumor, respectively. Tumor and nerve cell co-culture conditions were used to determine if myelinating or non-myelinating Schwann cell (S16 and S16Y, respectively) phenotype was equally likely to promote integrin-dependent cancer cell invasion and migration on laminin. Conditioned medium from S16 cells increased tumor cell (DU145, PC3, and CFPAC1) invasion into laminin approximately 1.3-2.0 fold compared to fetal bovine serum (FBS) treated cells. Integrin function (e.g., ITGA6p formation) increased up to 1.5 fold in prostate (DU145, PC3, RWPE-1) and pancreatic (CFPAC1) cells, and invasion was dependent on ITGA6p formation and ITGB1 as determined by function-blocking antibodies. In contrast, conditioned medium isolated from S16Y cells (non-myelinating phenotype) decreased constitutive levels of ITGA6p in the tumor cells by 50% compared to untreated cells and decreased ITGA6p formation 3.0 fold compared to S16 treated cells. Flow cytometry and western blot analysis revealed loss of ITGA6p formation as reversible and independent of overall loss of ITGA6 expression. These results suggest that the myelinating phenotype of Schwann cells within the tumor microenvironment increased integrin-dependent tumor invasion on laminin.
Collapse
Affiliation(s)
- Isis C Sroka
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona, 85724
| | - Harsharon Chopra
- Department of Pathology, University of Arizona College of Medicine, Tucson, Arizona, 85724
| | - Lipsa Das
- University of Arizona Cancer Center, 1515 North Campbell Avenue, Tucson, Arizona, 85724
| | - Jaime M C Gard
- University of Arizona Cancer Center, 1515 North Campbell Avenue, Tucson, Arizona, 85724
| | - Raymond B Nagle
- Department of Pathology, University of Arizona College of Medicine, Tucson, Arizona, 85724
| | - Anne E Cress
- University of Arizona Cancer Center, 1515 North Campbell Avenue, Tucson, Arizona, 85724.,Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona, 85724
| |
Collapse
|
25
|
Qin Y, Rodin S, Simonson OE, Hollande F. Laminins and cancer stem cells: Partners in crime? Semin Cancer Biol 2016; 45:3-12. [PMID: 27491691 DOI: 10.1016/j.semcancer.2016.07.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/30/2016] [Indexed: 01/31/2023]
Abstract
As one of the predominant protein families within the extracellular matrix both structurally and functionally, laminins have been shown to be heavily involved in tumor progression and drug resistance. Laminins participate in key cellular events for tumor angiogenesis, cell invasion and metastasis development, including the regulation of epithelial-mesenchymal transition and basement membrane remodeling, which are tightly associated with the phenotypic characteristics of stem-like cells, particularly in the context of cancer. In addition, a great deal of studies and reports has highlighted the critical roles of laminins in modulating stem cell phenotype and differentiation, as part of the stem cell niche. Stemming from these discoveries a growing body of literature suggests that laminins may act as regulators of cancer stem cells, a tumor cell subpopulation that plays an instrumental role in long-term cancer maintenance, metastasis development and therapeutic resistance. The accumulating evidence in this emerging research area suggests that laminins represent potential therapeutic targets for anti-cancer treatments against cancer stem cells, and that they may be used as predictive and prognostic markers to inform clinical management and improve patient survival.
Collapse
Affiliation(s)
- Yan Qin
- Department of Pathology, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Sergey Rodin
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.
| | - Oscar E Simonson
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden; Department of Cardiothoracic Surgery, Uppsala University Hospital, Uppsala, Sweden.
| | - Frédéric Hollande
- Department of Pathology, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
26
|
Brantley E, Callero MA, Berardi DE, Campbell P, Rowland L, Zylstra D, Amis L, Yee M, Simian M, Todaro L, Loaiza-Perez AI, Soto U. AhR ligand Aminoflavone inhibits α6-integrin expression and breast cancer sphere-initiating capacity. Cancer Lett 2016; 376:53-61. [PMID: 26996297 DOI: 10.1016/j.canlet.2016.03.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/13/2016] [Accepted: 03/14/2016] [Indexed: 01/25/2023]
Abstract
Traditional chemotherapies debulk tumors but fail to produce long-term clinical remissions due to their inability to eradicate tumor-initiating cells (TICs). This necessitates therapy with activity against the TIC niche. Αlpha6-integrin (α6-integrin) promotes TIC growth. In contrast, aryl hydrocarbon receptor (AhR) signaling activation impedes the formation of mammospheres (clusters of cells enriched for TICs). We investigated the ability of AhR agonist Aminoflavone (AF) and AF pro-drug (AFP464) to disrupt mammospheres derived from breast cancer cells and a M05 mammary mouse model of breast cancer respectively. We further examined the capacity of AF and AFP464 to exhibit anticancer activity and modulate the expression of 'stemness' genes including α6-integrin using immunofluorescence, flow cytometry and qRT-PCR analysis. AF disrupted mammospheres and prevented secondary mammosphere formation. In contrast, AF did not disrupt mammospheres derived from AhR ligand-unresponsive MCF-7 cells. AFP464 treatment suppressed M05 tumor growth and disrupted corresponding mammospheres. AF and AFP464 reduced the expression and percentage of cells that stained for 'stemness' markers including α6-integrin in vitro and in vivo respectively. These data suggest AFP464 thwarts bulk breast tumor and TIC growth via AhR agonist-mediated α6-integrin inhibition.
Collapse
Affiliation(s)
- Eileen Brantley
- Department of Basic Sciences, Loma Linda University Health School of Medicine, 11021 Campus St, Alumni Hall Room 101, Loma Linda, CA 92354, USA; Department of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA, USA
| | - Mariana A Callero
- Research Area, Institute of Oncology Ángel H. Roffo, University of Buenos Aires, Avenue San Martín 5481, C1417DTB Ciudad de Buenos Aires, Argentina
| | - Damian E Berardi
- Research Area, Institute of Oncology Ángel H. Roffo, University of Buenos Aires, Avenue San Martín 5481, C1417DTB Ciudad de Buenos Aires, Argentina
| | - Petreena Campbell
- Department of Basic Sciences, Loma Linda University Health School of Medicine, 11021 Campus St, Alumni Hall Room 101, Loma Linda, CA 92354, USA
| | - Leah Rowland
- Department of Basic Sciences, Loma Linda University Health School of Medicine, 11021 Campus St, Alumni Hall Room 101, Loma Linda, CA 92354, USA
| | - Dain Zylstra
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA, USA
| | - Louisa Amis
- Department of Basic Sciences, Loma Linda University Health School of Medicine, 11021 Campus St, Alumni Hall Room 101, Loma Linda, CA 92354, USA
| | - Michael Yee
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Marina Simian
- Research Area, Institute of Oncology Ángel H. Roffo, University of Buenos Aires, Avenue San Martín 5481, C1417DTB Ciudad de Buenos Aires, Argentina
| | - Laura Todaro
- Research Area, Institute of Oncology Ángel H. Roffo, University of Buenos Aires, Avenue San Martín 5481, C1417DTB Ciudad de Buenos Aires, Argentina
| | - Andrea I Loaiza-Perez
- Research Area, Institute of Oncology Ángel H. Roffo, University of Buenos Aires, Avenue San Martín 5481, C1417DTB Ciudad de Buenos Aires, Argentina.
| | - Ubaldo Soto
- Department of Basic Sciences, Loma Linda University Health School of Medicine, 11021 Campus St, Alumni Hall Room 101, Loma Linda, CA 92354, USA.
| |
Collapse
|
27
|
Brooks DLP, Schwab LP, Krutilina R, Parke DN, Sethuraman A, Hoogewijs D, Schörg A, Gotwald L, Fan M, Wenger RH, Seagroves TN. ITGA6 is directly regulated by hypoxia-inducible factors and enriches for cancer stem cell activity and invasion in metastatic breast cancer models. Mol Cancer 2016; 15:26. [PMID: 27001172 PMCID: PMC4802728 DOI: 10.1186/s12943-016-0510-x] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 03/11/2016] [Indexed: 11/27/2022] Open
Abstract
Background Hypoxia-inducible factors (HIFs) are well-established mediators of tumor growth, the epithelial to mesenchymal transition (EMT) and metastasis. In several types of solid tumors, including breast cancers, the HIFs play a critical role in maintaining cancer stem cell (CSC) activity. Thus, we hypothesized that HIFs may also regulate transcription of markers of breast CSC activity. One approach to enrich for breast cells with stem-like phenotypes is FACS sorting, in which sub-populations of live cells are gated based on the expression of cell surface antigens, including various integrin subunits. Integrin alpha 6 (ITGA6; CD49f) is routinely used in combination with other integrin subunits to enrich for breast stem cells by FACS. Integrins not only mediate interactions with the extracellular matrix (ECM), but also drive intracellular signaling events that communicate from the tumor microenvironment to inside of the tumor cell to alter phenotypes including migration and invasion. Methods We used two models of metastatic breast cancer (MBC), polyoma middle T (MMTV-PyMT) and MDA-MB-231 cells, to compare the expression of ITGA6 in wild type and knockout (KO) or knockdown cells. Chromatin immunoprecipitation (ChIP) and luciferase reporter assays verified that ITGA6 is a direct HIF transcriptional target. We also used FACS sorting to enrich for CD49f + cells to compare tumorsphere formation, tumor initiating cell activity, invasion and HIF activity relative to CD49fneg or low cells. Knockdown of ITGA6 significantly reduced invasion, whereas re-expression of ITGA6 in the context of HIF knockdown partially rescued invasion. A search of public databases also revealed that ITGA6 expression is an independent prognostic factor of survival in breast cancer patients. Results We report that ITGA6 is a HIF-dependent target gene and that high ITGA6 expression enhances invasion and tumor-initiating cell activities in models of MBC. Moreover, cells that express high levels of ITGA6 are enriched for HIF-1α expression and the expression of HIF-dependent target genes. Conclusions Our data suggest that HIF-dependent regulation of ITGA6 is one mechanism by which sorting for CD49f + cells enhances CSC and metastatic phenotypes in breast cancers. Our results are particularly relevant to basal-like breast cancers which express higher levels of the HIFα subunits, core HIF-dependent target genes and ITGA6 relative to other molecular subtypes. Electronic supplementary material The online version of this article (doi:10.1186/s12943-016-0510-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Danielle L Peacock Brooks
- Center for Cancer Research and the Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Present address: National Cancer Institute, Center for Cancer Research, Women's Malignancies Branch, Bethesda, MD, 20892, USA
| | - Luciana P Schwab
- Center for Cancer Research and the Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Raisa Krutilina
- Center for Cancer Research and the Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Deanna N Parke
- Center for Cancer Research and the Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Aarti Sethuraman
- Center for Cancer Research and the Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - David Hoogewijs
- Institute of Physiology and Zürich Center for Integrative Human Physiology, University of Zürich, CH-8057, Zürich, Switzerland.,Present address: Institute of Physiology, University of Duisburg-Essen, 45122, Essen, Germany
| | - Alexandra Schörg
- Institute of Physiology and Zürich Center for Integrative Human Physiology, University of Zürich, CH-8057, Zürich, Switzerland
| | - Lauren Gotwald
- Center for Cancer Research and the Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Meiyun Fan
- Center for Cancer Research and the Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Roland H Wenger
- Institute of Physiology and Zürich Center for Integrative Human Physiology, University of Zürich, CH-8057, Zürich, Switzerland
| | - Tiffany N Seagroves
- Center for Cancer Research and the Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
28
|
Harryman WL, Pond E, Singh P, Little AS, Eschbacher JM, Nagle RB, Cress AE. Laminin-binding integrin gene copy number alterations in distinct epithelial-type cancers. Am J Transl Res 2016; 8:940-954. [PMID: 27158381 PMCID: PMC4846938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/29/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND The laminin-binding integrin (LBI) family are cell adhesion molecules that are essential for invasion and metastasis of human epithelial cancers and cell adhesion mediated drug resistance. We investigated whether copy number alteration (CNA) or mutations of a five-gene signature (ITGB4, ITGA3, LAMB3, PLEC, and SYNE3), representing essential genes for LBI adhesion, would correlate with patient outcomes within human epithelial-type tumor data sets currently available in an open access format. METHODS We investigated the relative alteration frequency of an LBI signature panel (integrin β4 (ITGB4), integrin α3 (ITGA3), laminin β3 chain (LAMB3), plectin (PLEC), and nesprin 3 (SYNE3)), independent of the epithelial cancer type, within publically available and published data using cBioPortal and Oncomine software. We rank ordered the results using a 20% alteration frequency cut-off and limited the analysis to studies containing at least 100 samples. Kaplan-Meier survival curves were analyzed to determine if alterations in the LBI signature correlated with patient survival. The Oncomine data mining tool was used to compare the heat map expression of the LBI signature without SYNE3 (as this was not included in the Oncomine database) to drug resistance patterns. RESULTS Twelve different cancer types, representing 5,647 samples, contained at least a 20% alteration frequency of the five-gene LBI signature. The frequency of alteration ranged from 38.3% to 19.8%. Within the LBI signature, PLEC was the most commonly altered followed by LAMB3, ITGB4, ITGA3, and SYNE3 across all twelve cancer types. Within cancer types, there was little overlap of the individual amplified genes from each sample, suggesting different specific amplicons may alter the LBI adhesion structures. Of the twelve cancer types, overall survival was altered by CNA presence in bladder urothelial carcinoma (p=0.0143*) and cervical squamous cell carcinoma and endocervical adenocarcinoma (p=0.0432*). Querying the in vitro drug resistance profiles with the LBI signature demonstrated a positive correlation with cells resistant to inhibitors of HDAC (Vorinostat, Panobinostat) and topoisomerase II (Irinotecan). No correlation was found with the following agents: Bleomycin, Doxorubicin, Methotrexate, Gemcitabine, Docetaxel, Bortezomib, and Shikonen. CONCLUSIONS Our work has identified epithelial-types of human cancer that have significant CNA in our selected five-gene signature, which was based on the essential and genetically-defined functions of the protein product networks (in this case, the LBI axis). CNA of the gene signature not only predicted overall survival in bladder, cervical, and endocervical adenocarcinoma but also response to chemotherapy. This work suggests that future studies designed to optimize the gene signature are warranted. GENERAL SIGNIFICANCE The copy number alteration of structural components of the LBI axis in epithelial-type tumors may be promising biomarkers and rational targets for personalized therapy in preventing or arresting metastatic spread.
Collapse
Affiliation(s)
- William L Harryman
- The University of Arizona Cancer Center1515 N. Campbell Ave., Tucson, Arizona, United States
| | - Erika Pond
- The University of Arizona Cancer Center1515 N. Campbell Ave., Tucson, Arizona, United States
| | - Parminder Singh
- The University of Arizona Cancer Center1515 N. Campbell Ave., Tucson, Arizona, United States
| | - Andrew S Little
- Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center350 W. Thomas Rd., Phoenix, Arizona, United States
| | - Jennifer M Eschbacher
- Department of Pathology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center350 W. Thomas Rd., Phoenix, Arizona, United States
| | - Raymond B Nagle
- The University of Arizona Cancer Center1515 N. Campbell Ave., Tucson, Arizona, United States
| | - Anne E Cress
- The University of Arizona Cancer Center1515 N. Campbell Ave., Tucson, Arizona, United States
| |
Collapse
|
29
|
Ganguly SS, Li X, Miranti CK. The host microenvironment influences prostate cancer invasion, systemic spread, bone colonization, and osteoblastic metastasis. Front Oncol 2014; 4:364. [PMID: 25566502 PMCID: PMC4266028 DOI: 10.3389/fonc.2014.00364] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/29/2014] [Indexed: 12/28/2022] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer death in men worldwide. Most PCa deaths are due to osteoblastic bone metastases. What triggers PCa metastasis to the bone and what causes osteoblastic lesions remain unanswered. A major contributor to PCa metastasis is the host microenvironment. Here, we address how the primary tumor microenvironment influences PCa metastasis via integrins, extracellular proteases, and transient epithelia-mesenchymal transition (EMT) to promote PCa progression, invasion, and metastasis. We discuss how the bone-microenvironment influences metastasis; where chemotactic cytokines favor bone homing, adhesion molecules promote colonization, and bone-derived signals induce osteoblastic lesions. Animal models that fully recapitulate human PCa progression from primary tumor to bone metastasis are needed to understand the PCa pathophysiology that leads to bone metastasis. Better delineation of the specific processes involved in PCa bone metastasize is needed to prevent or treat metastatic PCa. Therapeutic regimens that focus on the tumor microenvironment could add to the PCa pharmacopeia.
Collapse
Affiliation(s)
- Sourik S Ganguly
- Program for Skeletal Disease and Tumor Metastasis, Laboratory of Tumor Microenvironment and Metastasis, Center for Cancer and Cell Biology, Van Andel Research Institute , Grand Rapids, MI , USA ; Program for Skeletal Disease and Tumor Metastasis, Laboratory of Integrin Signaling and Tumorigenesis, Center for Cancer and Cell Biology, Van Andel Research Institute , Grand Rapids, MI , USA
| | - Xiaohong Li
- Program for Skeletal Disease and Tumor Metastasis, Laboratory of Tumor Microenvironment and Metastasis, Center for Cancer and Cell Biology, Van Andel Research Institute , Grand Rapids, MI , USA
| | - Cindy K Miranti
- Program for Skeletal Disease and Tumor Metastasis, Laboratory of Integrin Signaling and Tumorigenesis, Center for Cancer and Cell Biology, Van Andel Research Institute , Grand Rapids, MI , USA
| |
Collapse
|
30
|
Mukherjee S, Saha S, Manna A, Mazumdar M, Chakraborty S, Paul S, Das T. Targeting Cancer Stem Cells by Phytochemicals: a Multimodal Approach to Colorectal Cancer. CURRENT COLORECTAL CANCER REPORTS 2014. [DOI: 10.1007/s11888-014-0251-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
31
|
Kacsinta AD, Rubenstein CS, Sroka IC, Pawar S, Gard JM, Nagle RB, Cress AE. Intracellular modifiers of integrin alpha 6p production in aggressive prostate and breast cancer cell lines. Biochem Biophys Res Commun 2014; 454:335-40. [PMID: 25450398 DOI: 10.1016/j.bbrc.2014.10.073] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/15/2014] [Indexed: 02/03/2023]
Abstract
Cancer metastasis is a multi-step process in which tumor cells gain the ability to invade beyond the primary tumor and colonize distant sites. The mechanisms regulating the metastatic process confer changes to cell adhesion receptors including the integrin family of receptors. Our group previously discovered that the α6 integrin (ITGA6/CD49f) is post translationally modified by urokinase plasminogen activator (uPA) and its receptor, urokinase plasminogen activator receptor (uPAR), to form the variant ITGA6p. This variant of ITGA6 is a cleaved form of the receptor that lacks the ligand-binding domain. Although it is established that the uPA/uPAR axis drives ITGA6 cleavage, the mechanisms regulating cleavage have not been defined. Intracellular integrin dependent "inside-out" signaling is a major regulator of integrin function and the uPA/uPAR axis. We hypothesized that intracellular signaling molecules play a role in formation of ITGA6p to promote cell migration during cancer metastasis. In order to test our hypothesis, DU145 and PC3B1 prostate cancer and MDA-MB-231 breast cancer cell lines were treated with small interfering RNA targeting actin and the intracellular signaling regulators focal adhesion kinase (FAK), integrin linked kinase (ILK), and paxillin. The results demonstrated that inhibition of actin, FAK, and ILK expression resulted in significantly increased uPAR expression and ITGA6p production. Inhibition of actin increased ITGA6p, although inhibition of paxillin did not affect ITGA6p formation. Taken together, these results suggest that FAK and ILK dependent "inside-out" signaling, and actin dynamics regulate extracellular production of ITGA6p and the aggressive phenotype.
Collapse
Affiliation(s)
- Apollo D Kacsinta
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States; Cancer Biology Interdisciplinary Graduate Program, University of Arizona, Tucson, AZ, United States
| | - Cynthia S Rubenstein
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States; Cancer Biology Interdisciplinary Graduate Program, University of Arizona, Tucson, AZ, United States
| | - Isis C Sroka
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States; Cancer Biology Interdisciplinary Graduate Program, University of Arizona, Tucson, AZ, United States
| | - Sangita Pawar
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States; Department of Pharmacology, University of Arizona, Tucson, AZ, United States
| | - Jaime M Gard
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
| | - Raymond B Nagle
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States; Department of Pathology, University of Arizona, Tucson, AZ, United States
| | - Anne E Cress
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States; Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States.
| |
Collapse
|