1
|
Jiang L, Khawaja H, Tahsin S, Clarkson TA, Miranti CK, Zohar Y. Microfluidic-based human prostate-cancer-on-chip. Front Bioeng Biotechnol 2024; 12:1302223. [PMID: 38322789 PMCID: PMC10844564 DOI: 10.3389/fbioe.2024.1302223] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024] Open
Abstract
Lack of adequate models significantly hinders advances in prostate cancer treatment, where resistance to androgen-deprivation therapies and bone metastasis remain as major challenges. Current in vitro models fail to faithfully mimic the complex prostate physiology. In vivo animal models can shed light on the oncogenes involved in prostate cancer development and progression; however, the animal prostate gland is fundamentally different from that of human, and the underlying genetic mechanisms are different. To address this problem, we developed the first in vitro microfluidic human Prostate-Cancer-on-Chip (PCoC) model, where human prostate cancer and stromal fibroblast cells were co-cultivated in two channels separated by a porous membrane under culture medium flow. The established microenvironment enables soluble signaling factors secreted by each culture to locally diffuse through the membrane pores affecting the neighboring culture. We particularly explored the conversion of the stromal fibroblasts into cancer-associated fibroblasts (CAFs) due to the interaction between the 2 cell types. Immunofluorescence microscopy revealed that tumor cells induced CAF biomarkers, αSMA and COL1A1, in stromal fibroblasts. The stromal CAF conversion level was observed to increase along the flow direction in response to diffusion agents, consistent with simulations of solute concentration gradients. The tumor cells also downregulated androgen receptor (AR) expression in stromal fibroblasts, while an adequate level of stromal AR expression is maintained in normal prostate homeostasis. We further investigated tumor invasion into the stroma, an early step in the metastatic cascade, in devices featuring a serpentine channel with orthogonal channel segments overlaying a straight channel and separated by an 8 µm-pore membrane. Both tumor cells and stromal CAFs were observed to cross over into their neighboring channel, and the stroma's role seemed to be proactive in promoting cell invasion. As control, normal epithelial cells neither induced CAF conversion nor promoted cell invasion. In summary, the developed PCoC model allows spatiotemporal analysis of the tumor-stroma dynamic interactions, due to bi-directional signaling and physical contact, recapitulating tissue-level multicellular responses associated with prostate cancer in vivo. Hence, it can serve as an in vitro model to dissect mechanisms in human prostate cancer development and seek advanced therapeutic strategies.
Collapse
Affiliation(s)
- Linan Jiang
- Department of Aerospace and Mechanical Engineering, Tucson, AZ, United States
| | - Hunain Khawaja
- Cancer Biology Graduate Interdisciplinary Program, Tucson, AZ, United States
| | - Shekha Tahsin
- Cancer Biology Graduate Interdisciplinary Program, Tucson, AZ, United States
| | | | - Cindy K. Miranti
- Department of Molecular and Cellular Biology, Tucson, AZ, United States
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, Tucson, AZ, United States
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
2
|
Wolf I, Gratzke C, Wolf P. Prostate Cancer Stem Cells: Clinical Aspects and Targeted Therapies. Front Oncol 2022; 12:935715. [PMID: 35875084 PMCID: PMC9304860 DOI: 10.3389/fonc.2022.935715] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Despite decades of research and successful improvements in diagnosis and therapy, prostate cancer (PC) remains a major challenge. In recent years, it has become clear that PC stem cells (PCSCs) are the driving force in tumorigenesis, relapse, metastasis, and therapeutic resistance of PC. In this minireview, we discuss the impact of PCSCs in the clinical practice. Moreover, new therapeutic approaches to combat PCSCs are presented with the aim to achieve an improved outcome for patients with PC.
Collapse
Affiliation(s)
- Isis Wolf
- Department of Urology, Medical Center-University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Gratzke
- Department of Urology, Medical Center-University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Wolf
- Department of Urology, Medical Center-University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- *Correspondence: Philipp Wolf,
| |
Collapse
|
3
|
Transcriptomic Analysis of LNCaP Tumor Xenograft to Elucidate the Components and Mechanisms Contributed by Tumor Environment as Targets for Dietary Prostate Cancer Prevention Studies. Nutrients 2021; 13:nu13031000. [PMID: 33808801 PMCID: PMC8003580 DOI: 10.3390/nu13031000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 11/24/2022] Open
Abstract
LNCaP athymic xenograft model has been widely used to allow researchers to examine the effects and mechanisms of experimental treatments such as diet and diet-derived cancer preventive and therapeutic compounds on prostate cancer. However, the biological characteristics of human LNCaP cells before/after implanting in athymic mouse and its relevance to clinical human prostate outcomes remain unclear and may dictate interpretation of biological efficacies/mechanisms of diet/diet-derived experimental treatments. In this study, transcriptome profiles and pathways of human prostate LNCaP cells before (in vitro) and after (in vivo) implanting into xenograft mouse were compared using RNA-sequencing technology (RNA-seq) followed by bioinformatic analysis. A shift from androgen-responsive to androgen nonresponsive status was observed when comparing LNCaP xenograft tumor to culture cells. Androgen receptor and aryl-hydrocarbon pathway were found to be inhibited and interleukin-1 (IL-1) mediated pathways contributed to these changes. Coupled with in vitro experiments modeling for androgen exposure, cell-matrix interaction, inflammation, and hypoxia, we identified specific mechanisms that may contribute to the observed changes in genes and pathways. Our results provide critical baseline transcriptomic information for a tumor xenograft model and the tumor environments that might be associated with regulating the progression of the xenograft tumor, which may influence interpretation of diet/diet-derived experimental treatments.
Collapse
|
4
|
Shekari F, Han CL, Lee J, Mirzaei M, Gupta V, Haynes PA, Lee B, Baharvand H, Chen YJ, Hosseini Salekdeh G. Surface markers of human embryonic stem cells: a meta analysis of membrane proteomics reports. Expert Rev Proteomics 2018; 15:911-922. [PMID: 30358457 DOI: 10.1080/14789450.2018.1539669] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/19/2018] [Indexed: 12/12/2022]
Abstract
Human embryonic stem cells (hESCs) have unique biological features and attributes that make them attractive in various areas of biomedical research. With heightened applications, there is an ever increasing need for advancement of proteome analysis. Membrane proteins are one of the most important subset of hESC proteins as they can be used as surface markers. Areas covered: This review discusses commonly used surface markers of hESCs, and provides in-depth analysis of available hESC membrane proteome reports and the existence of these markers in many other cell types, especially cancer cells. Appreciating, existing ambiguity in the definition of a membrane protein, we have attempted a meta analysis of the published membrane protein reports of hESCs by using a combination of protein databases and prediction tools to find the most confident plasma membrane proteins in hESCs. Furthermore, responsiveness of plasma membrane proteins to differentiation has been discussed based on available transcriptome profiling data bank. Expert commentary: Combined transcriptome and membrane proteome analysis highlighted additional proteins that may eventually find utility as new cell surface markers.
Collapse
Affiliation(s)
- Faezeh Shekari
- a Department of Molecular Systems Biology at Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
- b Department of Developmental Biology , University of Science and Culture, ACECR , Tehran , Iran
| | - Chia-Li Han
- c Chemical Biology and Molecular Biophysics Program , Institute of Chemistry , Taipei , Taiwan , Republic of China
| | - Jaesuk Lee
- d Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute , Gachon University , Incheon , Republic of Korea
| | - Mehdi Mirzaei
- e Department of Molecular Sciences , Macquarie University , Sydney , NSW , Australia
- f Australian Proteome Analysis Facility , Macquarie University , Sydney , NSW , Australia
- g Department of Clinical Medicine , Macquarie University , Sydney , NSW , Australia
| | - Vivek Gupta
- g Department of Clinical Medicine , Macquarie University , Sydney , NSW , Australia
| | - Paul A Haynes
- e Department of Molecular Sciences , Macquarie University , Sydney , NSW , Australia
| | - Bonghee Lee
- d Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute , Gachon University , Incheon , Republic of Korea
| | - Hossein Baharvand
- b Department of Developmental Biology , University of Science and Culture, ACECR , Tehran , Iran
- h Department of Stem Cells and Developmental Biology at Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
| | - Yu-Ju Chen
- c Chemical Biology and Molecular Biophysics Program , Institute of Chemistry , Taipei , Taiwan , Republic of China
| | - Ghasem Hosseini Salekdeh
- a Department of Molecular Systems Biology at Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
- e Department of Molecular Sciences , Macquarie University , Sydney , NSW , Australia
- i Department of Systems and Synthetic biology , Agricultural Biotechnology Research Institute of Iran (ABRII), Agricultural Research, Education, and Extension Organization , Karaj , Iran
| |
Collapse
|
5
|
Meta-analysis of gene expression and integrin-associated signaling pathways in papillary renal cell carcinoma subtypes. Oncotarget 2018; 7:84178-84189. [PMID: 27705936 PMCID: PMC5356653 DOI: 10.18632/oncotarget.12390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 09/24/2016] [Indexed: 12/02/2022] Open
Abstract
Papillary renal cell carcinoma (PRCC) is the second most common renal cell carcinoma (RCC) that can be further subdivided into type 1 (PRCC1) and type 2 (PRCC2) RCCs based on histological and genetic features. PRCC2 is often more aggressive than PRCC1. While integrin-associated protein complexes mediate tumorigenesis and metastases in many types of cancers it is not known whether integrin-mediated signaling impacts PRCC and differs between PRCC1 and PRCC2. In this study, we combined the analysis of five PRCC gene expression datasets derived from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) by using integrative bioinformatics pipelines. We found 1475 differentially expressed genes among which 37 genes were associated with integrin pathways. In comparison with PRCC1, PRCC2 cases showed upregulated expression of α5-integrin (ITGA5) whereas the expression of α6- (ITGA6) and β8-integrins (ITGB8) was downregulated. Because PRCC2 occurs more frequently in men, the meta-analysis was extended to explore the gender effects. This analysis revealed 8 genes but none of them was related to integrin pathways suggesting that other mechanisms than integrin-mediated signaling underlie the observed gender differences in the pathogenicity of PRCC2.
Collapse
|
6
|
Groulx JF, Boudjadi S, Beaulieu JF. MYC Regulates α6 Integrin Subunit Expression and Splicing Under Its Pro-Proliferative ITGA6A Form in Colorectal Cancer Cells. Cancers (Basel) 2018; 10:42. [PMID: 29401653 PMCID: PMC5836074 DOI: 10.3390/cancers10020042] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 01/29/2018] [Accepted: 01/31/2018] [Indexed: 12/15/2022] Open
Abstract
The α6 integrin subunit (ITGA6) pre-mRNA undergoes alternative splicing to form two splicing variants, named ITGA6A and ITGA6B. In primary human colorectal cancer cells, the levels of both ITGA6 and β4 integrin subunit (ITGB4) subunits of the α6β4 integrin are increased. We previously found that the upregulation of ITGA6 is a direct consequence of the increase of the pro-proliferative ITGA6A variant. However, the mechanisms that control ITGA6 expression and splicing into the ITGA6A variant over ITGA6B in colorectal cancer cells remain poorly understood. Here, we show that the promoter activity of the ITGA6 gene is regulated by MYC. Pharmacological inhibition of MYC activity with the MYC inhibitor (MYCi) 10058-F4 or knockdown of MYC expression by short hairpin RNA (shRNA) both lead to a decrease in ITGA6 and ITGA6A levels in colorectal cancer cells, while overexpression of MYC enhances ITGA6 promoter activity. We also found that MYC inhibition decreases the epithelial splicing regulatory protein 2 (ESRP2) splicing factor at both the mRNA and protein levels. Chromatin immunoprecipitation revealed that the proximal promoter sequences of ITGA6 and ESRP2 were occupied by MYC and actively transcribed in colorectal cancer cells. Furthermore, expression studies in primary colorectal tumors and corresponding resection margins confirmed that the up-regulation of the ITGA6A subunit can be correlated with the increase in MYC and ESRP2. Taken together, our results demonstrate that the proto-oncogene MYC can regulate the promoter activation and splicing of the ITGA6 integrin gene through ESRP2 to favor the production of the pro-proliferative ITGA6A variant in colorectal cancer cells.
Collapse
Affiliation(s)
- Jean-François Groulx
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Salah Boudjadi
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
- Laboratory of Pathology, Cancer Molecular Pathology Section, National Cancer Institute, Bethesda, MD 20892, USA.
| | - Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| |
Collapse
|
7
|
Anandi L, Chakravarty V, Ashiq KA, Bodakuntla S, Lahiri M. DNA-dependent protein kinase plays a central role in transformation of breast epithelial cells following alkylation damage. J Cell Sci 2017; 130:3749-3763. [PMID: 28923836 DOI: 10.1242/jcs.203034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 09/14/2017] [Indexed: 12/25/2022] Open
Abstract
DNA alkylating agents form the first line of cancer chemotherapy. They not only kill cells but also behave as potential carcinogens. MNU, a DNA methylating agent, is well known to induce mammary tumours in rodents. However, the mechanism of tumorigenesis is not well understood. Our study reports a novel role played by DNA-dependent protein kinase (DNA-PK) in methylation damage-induced transformation using three-dimensional breast acinar cultures. Here, we report that exposure of breast epithelial cells to MNU inhibited polarisation at the basolateral domain, increased dispersal of the Golgi at the apical domain and induced an epithelial-to-mesenchymal transition (EMT)-like phenotype as well as invasion. This altered Golgi phenotype correlated with impaired intracellular trafficking. Inhibition of DNA-PK resulted in almost complete reversal of the altered Golgi phenotype and partial rescue of the polarity defect and EMT-like phenotype. The results confirm that methylation damage-induced activation of DNA-PK is a major mechanism in mediating cellular transformation.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Libi Anandi
- Indian Institute of Science Education and Research, Pune, Maharashtra 411008, India
| | - Vaishali Chakravarty
- Indian Institute of Science Education and Research, Pune, Maharashtra 411008, India
| | - K A Ashiq
- Indian Institute of Science Education and Research, Pune, Maharashtra 411008, India
| | - Satish Bodakuntla
- Indian Institute of Science Education and Research, Pune, Maharashtra 411008, India.,Institut Curie, PSL Research University, CNRS UMR3348, F-91405 Orsay, France
| | - Mayurika Lahiri
- Indian Institute of Science Education and Research, Pune, Maharashtra 411008, India
| |
Collapse
|
8
|
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: 32] [Impact Index Per Article: 3.6] [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
|
9
|
The expression pattern and functional role of REIC/Dkk-3 in the development of cutaneous squamous cell carcinoma. J Dermatol Sci 2016; 84:88-96. [DOI: 10.1016/j.jdermsci.2016.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 12/30/2022]
|
10
|
Levine RM, Dinh CV, Harris MA, Kokkoli E. Targeting HPV-infected cervical cancer cells with PEGylated liposomes encapsulating siRNA and the role of siRNA complexation with polyethylenimine. Bioeng Transl Med 2016; 1:168-180. [PMID: 29313012 PMCID: PMC5675078 DOI: 10.1002/btm2.10022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 07/14/2016] [Accepted: 07/15/2016] [Indexed: 01/02/2023] Open
Abstract
The greatest obstacle to clinical application of cancer gene therapy is lack of effective delivery tools. Gene delivery vehicles must protect against degradation, avoid immunogenic effects and prevent off target delivery which can cause harmful side effects. PEGylated liposomes have greatly improved tumor localization of small molecule drugs and are a promising tool for nucleic acid delivery as the polyethylene glycol (PEG) coating protects against immune recognition and blood clearance. In this study, small interfering RNA (siRNA) was fully encapsulated within PEGylated liposomes by complexing the siRNA with a cationic polymer, polyethyleneimine (PEI), before encapsulation. Formation methods and material compositions were then investigated for their effects on encapsulation. This technology was translated for protective delivery of siRNA designed for human papillomavirus (HPV) viral gene silencing and cervical cancer treatment. PEGylated liposomes encapsulating siRNA were functionalized with the AG86 targeting peptide-amphiphile which binds to the α6β4 integrin, a cervical cancer biomarker. It was found that both targeting and polymer complexation before encapsulation were critical components to effective transfection.
Collapse
Affiliation(s)
- Rachel M. Levine
- Dept. of Chemical Engineering and Materials ScienceUniversity of MinnesotaMinneapolisMN55455
| | - Christina V. Dinh
- Dept. of Chemical Engineering and Materials ScienceUniversity of MinnesotaMinneapolisMN55455
| | - Michael A. Harris
- Dept. of Chemical Engineering and Materials ScienceUniversity of MinnesotaMinneapolisMN55455
| | - Efrosini Kokkoli
- Dept. of Chemical Engineering and Materials ScienceUniversity of MinnesotaMinneapolisMN55455
| |
Collapse
|
11
|
Fahey JM, Girotti AW. Accelerated migration and invasion of prostate cancer cells after a photodynamic therapy-like challenge: Role of nitric oxide. Nitric Oxide 2015; 49:47-55. [PMID: 26068242 DOI: 10.1016/j.niox.2015.05.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/14/2015] [Accepted: 05/26/2015] [Indexed: 11/25/2022]
Abstract
Employing an in vitro model for 5-aminolevulinic acid (ALA)-based photodynamic therapy (PDT), we recently reported that human prostate cancer PC3 cells rapidly and persistently overexpressed inducible nitric oxide synthase (iNOS) and nitric oxide (NO) after a moderate ALA/light challenge. The upregulated iNOS/NO was shown to play a key role in cell resistance to apoptotic photokilling and also in the dramatic growth spurt observed in surviving cells. In the present study, we found that PC3 cells surviving an ALA/light insult not only proliferated faster than non-stressed controls, but migrated and invaded faster as well, these effects being abrogated by an iNOS inhibitor or NO scavenger. Photostressed prostate DU145 cells exhibited similar behavior. Using in-gel zymography, we showed that PC3 extracellular matrix metalloproteinase-9 (MMP-9) was strongly activated 24 h after ALA/light treatment and that MMP-9 inhibitor TIMP-1 was downregulated, consistent with MMP-9 involvement in enhanced invasiveness. We also observed a photostress-induced upregulation of α6 and β1 integrins, implying their involvement as well. The MMP-9, TIMP-1, and integrin effects were strongly attenuated by iNOS inhibition, confirming NO's role in photostress-enhanced migration/invasion. This study reveals novel, potentially tumor-promoting, side-effects of prostate cancer PDT which may be averted through use of iNOS inhibitors as PDT adjuvants.
Collapse
Affiliation(s)
- Jonathan M Fahey
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226-3548, USA
| | - Albert W Girotti
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226-3548, USA.
| |
Collapse
|
12
|
Smadja DM, Guerin CL, Boscolo E, Bieche I, Mulliken JB, Bischoff J. α6-Integrin is required for the adhesion and vasculogenic potential of hemangioma stem cells. Stem Cells 2014; 32:684-93. [PMID: 24022922 DOI: 10.1002/stem.1539] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 07/31/2013] [Accepted: 08/12/2013] [Indexed: 11/10/2022]
Abstract
Infantile hemangioma (IH) is the most common tumor of infancy. Hemangioma stem cells (HemSC) are a mesenchymal subpopulation isolated from IH CD133+ cells. HemSC can differentiate into endothelial and pericyte/smooth muscle cells and form vascular networks when injected in immune-deficient mice. α6-Integrin subunit has been implicated in the tumorgenicity of glioblastoma stem cells and the homing properties of hematopoietic, endothelial, and mesenchymal progenitor cells. Therefore, we investigated the possible function(s) of α6-integrin in HemSC. We documented α6-integrin expression in IH tumor specimens and HemSC by RT-qPCR and flow cytometry. We examined the effect of blocking or silencing α6-integrin on the adhesive and proliferative properties of HemSC in vitro and the vasculogenic and homing properties of HemSC in vivo. Targeting α6-integrin in cultured HemSC inhibited adhesion to laminin but had no effect on proliferation. Vessel-forming ability in Matrigel implants and hepatic homing after i.v. delivery were significantly decreased in α6-integrin siRNA-transfected HemSC. In conclusion, α6-integrin is required for HemSC adherence to laminin, vessel formation in vivo, and for homing to the liver. Thus, we uncovered an important role for α6 integrin in the vasculogenic properties of HemSC. Our results suggest that α6-integrin expression on HemSC could be a new target for antihemangioma therapy.
Collapse
Affiliation(s)
- David M Smadja
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA; Paris Descartes University, Sorbonne Paris Cite, Paris, France; AP-HP, Hôpital Européen Georges Pompidou, Department of Hematology, Paris, France
| | | | | | | | | | | |
Collapse
|
13
|
Bouvard C, Segaoula Z, De Arcangelis A, Galy-Fauroux I, Mauge L, Fischer AM, Georges-Labouesse E, Helley D. Tie2-dependent deletion of α6 integrin subunit in mice reduces tumor growth and angiogenesis. Int J Oncol 2014; 45:2058-64. [PMID: 25176420 DOI: 10.3892/ijo.2014.2631] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 07/17/2014] [Indexed: 11/05/2022] Open
Abstract
The α6 integrin subunit (α6) has been implicated in cancer cell migration and in the progression of several malignancies, but its role in tumor angiogenesis is unclear. In mice, anti-α6 blocking antibodies reduce tumor angiogenesis, whereas Tie1-dependent α6 gene deletion enhances neovessel formation in melanoma and lung carcinoma. To clarify the discrepancy in these results we used the cre-lox system to generate a mouse line, α6fl/fl‑Tie2Cre(+), with α6 gene deletion specifically in Tie2-lineage cells: endothelial cells, pericytes, subsets of hematopoietic stem cells, and Tie2-expressing monocytes/macrophages (TEMs), known for their proangiogenic properties. Loss of α6 expression in α6fl/fl‑Tie2Cre(+) mice reduced tumor growth in a murine B16F10 melanoma model. Immunohistological analysis of the tumors showed that Tie2-dependent α6 gene deletion was associated with reduced tumor vascularization and with reduced infiltration of proangiogenic Tie2-expressing macrophages. These findings demonstrate that α6 integrin subunit plays a major role in tumor angiogenesis and TEM infiltration. Targeting α6 could be used as a strategy to reduce tumor growth.
Collapse
Affiliation(s)
| | | | - Adèle De Arcangelis
- Institute of Genetics, Cellular and Molecular Biology, INSERM U964, CNRS UMR 7104, University of Strasbourg, Illkirch, France
| | | | - Laetitia Mauge
- University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Elisabeth Georges-Labouesse
- Institute of Genetics, Cellular and Molecular Biology, INSERM U964, CNRS UMR 7104, University of Strasbourg, Illkirch, France
| | | |
Collapse
|
14
|
PGE2 signaling and its biosynthesis-related enzymes in cholangiocarcinoma progression. Tumour Biol 2014; 35:8051-64. [PMID: 24839005 DOI: 10.1007/s13277-014-2021-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 04/27/2014] [Indexed: 12/19/2022] Open
Abstract
Prostaglandin E2 (PGE2) involves in progression of various chronic inflammation-related cancers including cholangiocarcinoma (CCA). This study aimed to determine the role of PGE2 signaling, its biosynthesis-related enzymes in a clinical prognosis, and their targeted inhibition in CCA progression. The immunohistochemical staining of cyclooxygenase (COX)-1, COX-2, mPGES-1, EP1, and EP4 was examined in CCA tissues, and their expressions were compared with clinicopathological parameters. The effect of PGE2 on levels of its signaling molecules was examined in CCA cell lines using proteome profiler array. The suppression of mPGES-1 using a small-molecule inhibitor (CAY10526) and small interfering RNA (siRNA) was determined for growth and migration ability in CCA cells. The results indicated that strong expressions of COX-1, COX-2, mPGES-1, EP1, and EP4 were found in CCA tissues as 87.5, 47.5, 52.5, 55, and 80 % of frequencies, respectively. High mPGES-1 expression was significantly correlated with tumor stages III-IV (p = 0.001), lymph node metastasis (p = 0.004), shorter survival (p = 0.009), and prognostic indicator of CCA patients (HR = 2.512, p = 0.041). Expressions of COX-1, COX-2, and EP receptors did not correlate with data tested from patients. PGE2 markedly enhanced protein levels of integrinα6, VE-cadherin, Jagged1, and Notch3, and CAY10526 suppressed those protein levels as well as PGE2 production in CCA cells. CAY10526 and siRNA mPGES-1 markedly suppressed mPGES-1 protein levels, growth, and migration abilities of CCA cell lines. In conclusion, PGE2 signaling strongly promotes CCA progression. Therefore, inhibition of PGE2 synthesis by suppression of its biosynthesis-related enzymes could be useful for prevention and treatment of CCA.
Collapse
|
15
|
Menko AS, Bleaken BM, Libowitz AA, Zhang L, Stepp MA, Walker JL. A central role for vimentin in regulating repair function during healing of the lens epithelium. Mol Biol Cell 2014; 25:776-90. [PMID: 24478454 PMCID: PMC3952848 DOI: 10.1091/mbc.e12-12-0900] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A unique ex vivo mock cataract surgery model is used to study the role of vimentin in repair cell function during wound healing within a clinically relevant setting. Vimentin is found to be critical for the function of repair cells in directing the collective migration of the epithelium during wound healing. Mock cataract surgery provides a unique ex vivo model for studying wound repair in a clinically relevant setting. Here wound healing involves a classical collective migration of the lens epithelium, directed at the leading edge by an innate mesenchymal subpopulation of vimentin-rich repair cells. We report that vimentin is essential to the function of repair cells as the directors of the wound-healing process. Vimentin and not actin filaments are the predominant cytoskeletal elements in the lamellipodial extensions of the repair cells at the wound edge. These vimentin filaments link to paxillin-containing focal adhesions at the lamellipodial tips. Microtubules are involved in the extension of vimentin filaments in repair cells, the elaboration of vimentin-rich protrusions, and wound closure. The requirement for vimentin in repair cell function is revealed by both small interfering RNA vimentin knockdown and exposure to the vimentin-targeted drug withaferin A. Perturbation of vimentin impairs repair cell function and wound closure. Coimmunoprecipitation analysis reveals for the first time that myosin IIB is associated with vimentin, linking vimentin function in cell migration to myosin II motor proteins. These studies reveal a critical role for vimentin in repair cell function in regulating the collective movement of the epithelium in response to wounding.
Collapse
Affiliation(s)
- A S Menko
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107 Wills Vision Research Center at Jefferson, Philadelphia, PA 19107 Department of Anatomy and Regenerative Biology, George Washington University, Washington, DC 20037
| | | | | | | | | | | |
Collapse
|
16
|
Isaac J, Tarapore P, Zhang X, Lam YW, Ho SM. Site-specific S-nitrosylation of integrin α6 increases the extent of prostate cancer cell migration by enhancing integrin β1 association and weakening adherence to laminin-1. Biochemistry 2012; 51:9689-97. [PMID: 23106339 DOI: 10.1021/bi3012324] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The increased mortality in prostate cancer is usually the result of metastatic progression of the disease from the organ-confined location. Among the major events in this progression cascade are enhanced cell migration and loss of adhesion. Moreover, elevated levels of nitric oxide (NO) and inducible nitric oxide synthase (iNOS) found within the tumor microenvironment are hallmarks of progression of this cancer. To understand the role of nitrosative stress in prostate cancer progression, we investigated the effects of NO and iNOS on prostate cancer cell migration and adhesion. Our results indicate that ectopic expression of iNOS in prostate cancer cells increased the extent of cell migration, which could be blocked by selective ITGα6 blocking antibody or iNOS inhibitors. Furthermore, iNOS was found to cause S-nitrosylation of ITGα6 at Cys86 in prostate cancer cells. By comparing the activities of wild-type ITGα6 and a Cys86 mutant, we showed that treatment of prostate cancer cells with NO increased the level of ITGα6 heterodimerization with ITGβ1 but not with ITGβ4. Finally, S-nitrosylation of ITGα6 weakened its binding to laminin-β1 and weakened the adhesion of prostate cancer cells to laminin-1. In conclusion, S-nitrosylation of ITGα6 increased the extent of prostate cancer cell migration, which could be a potential mechanism of NO- and iNOS-induced enhancement of prostate cancer metastasis.
Collapse
Affiliation(s)
- Jared Isaac
- Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | | | | | |
Collapse
|
17
|
RGD-Binding Integrins in Prostate Cancer: Expression Patterns and Therapeutic Prospects against Bone Metastasis. Cancers (Basel) 2012; 4:1106-45. [PMID: 24213501 PMCID: PMC3712721 DOI: 10.3390/cancers4041106] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 10/09/2012] [Accepted: 10/22/2012] [Indexed: 12/26/2022] Open
Abstract
Prostate cancer is the third leading cause of male cancer deaths in the developed world. The current lack of highly specific detection methods and efficient therapeutic agents for advanced disease have been identified as problems requiring further research. The integrins play a vital role in the cross-talk between the cell and extracellular matrix, enhancing the growth, migration, invasion and metastasis of cancer cells. Progression and metastasis of prostate adenocarcinoma is strongly associated with changes in integrin expression, notably abnormal expression and activation of the β3 integrins in tumour cells, which promotes haematogenous spread and tumour growth in bone. As such, influencing integrin cell expression and function using targeted therapeutics represents a potential treatment for bone metastasis, the most common and debilitating complication of advanced prostate cancer. In this review, we highlight the multiple ways in which RGD-binding integrins contribute to prostate cancer progression and metastasis, and identify the rationale for development of multi-integrin antagonists targeting the RGD-binding subfamily as molecularly targeted agents for its treatment.
Collapse
|
18
|
Gene expression profiling and pathway analysis identify the integrin signaling pathway to be altered by IL-1β in human pancreatic cancer cells: Role of JNK. Cancer Lett 2012; 320:86-95. [DOI: 10.1016/j.canlet.2012.01.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 01/18/2012] [Accepted: 01/25/2012] [Indexed: 11/23/2022]
|
19
|
Zhao W, Ji X, Zhang F, Li L, Ma L. Embryonic stem cell markers. Molecules 2012; 17:6196-236. [PMID: 22634835 PMCID: PMC6268870 DOI: 10.3390/molecules17066196] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 03/31/2012] [Accepted: 05/04/2012] [Indexed: 02/07/2023] Open
Abstract
Embryonic stem cell (ESC) markers are molecules specifically expressed in ES cells. Understanding of the functions of these markers is critical for characterization and elucidation for the mechanism of ESC pluripotent maintenance and self-renewal, therefore helping to accelerate the clinical application of ES cells. Unfortunately, different cell types can share single or sometimes multiple markers; thus the main obstacle in the clinical application of ESC is to purify ES cells from other types of cells, especially tumor cells. Currently, the marker-based flow cytometry (FCM) technique and magnetic cell sorting (MACS) are the most effective cell isolating methods, and a detailed maker list will help to initially identify, as well as isolate ESCs using these methods. In the current review, we discuss a wide range of cell surface and generic molecular markers that are indicative of the undifferentiated ESCs. Other types of molecules, such as lectins and peptides, which bind to ESC via affinity and specificity, are also summarized. In addition, we review several markers that overlap with tumor stem cells (TSCs), which suggest that uncertainty still exists regarding the benefits of using these markers alone or in various combinations when identifying and isolating cells.
Collapse
Affiliation(s)
- Wenxiu Zhao
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
| | - Xiang Ji
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China
| | - Fangfang Zhang
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China
| | - Liang Li
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China
| | - Lan Ma
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
| |
Collapse
|
20
|
Bouvard C, De Arcangelis A, Dizier B, Galy-Fauroux I, Fischer AM, Georges-Labouesse E, Helley D. Tie2-dependent knockout of α6 integrin subunit in mice reduces post-ischaemic angiogenesis. Cardiovasc Res 2012; 95:39-47. [DOI: 10.1093/cvr/cvs153] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
21
|
Frick A, Grammel D, Schmidt F, Pöschl J, Priller M, Pagella P, von Bueren AO, Peraud A, Tonn JC, Herms J, Rutkowski S, Kretzschmar HA, Schüller U. Proper cerebellar development requires expression of β1-integrin in Bergmann glia, but not in granule neurons. Glia 2012; 60:820-32. [PMID: 22374686 DOI: 10.1002/glia.22314] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 02/01/2012] [Indexed: 01/20/2023]
Abstract
β1-class integrins play essential roles both in developmental biology as well as in cancer. Particularly, a Nestin-driven deletion of β1-integrin receptors results in severe abnormalities of brain development including a laminar disorganization of cerebellar granule neurons. However, since Nestin is expressed in all kinds of neural precursors, these data do not allow conclusions to be drawn about the role of β1-integrins in distinct neuronal and glial cell types. By generating conditional knockout mice using granule cell-specific Math1-promoter sequences, we show here that the expression of β1-integrins in granule neurons is dispensable for the development of the cerebellum. Also, deletion of β1-integrin from tumors that arise in a mouse model of granule cell precursor-derived medulloblastoma did not result in a significant survival benefit. Last, expression levels of β1-integrin in human medulloblastoma samples did not predict patient's outcome. However, a β1-integrin knockout using hGFAP-promoter sequences led to cerebellar hypoplasia, inappropriate positioning of Bergmann glia cells in the molecular layer, undirected outgrowth of radial glia fibers, and granule cell ectopia. We therefore conclude that β1-integrin expression in cerebellar granule neurons is not essential during normal development or medulloblastoma formation. In fact, it is the expression of β1-integrin in glia that is crucial for the proper development of the cerebellar cortex.
Collapse
Affiliation(s)
- Alexandra Frick
- Center for Neuropathology, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Castilla C, Flores ML, Conde JM, Medina R, Torrubia FJ, Japón MA, Sáez C. Downregulation of protein tyrosine phosphatase PTPL1 alters cell cycle and upregulates invasion-related genes in prostate cancer cells. Clin Exp Metastasis 2012; 29:349-58. [PMID: 22274591 DOI: 10.1007/s10585-012-9455-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 01/13/2012] [Indexed: 12/30/2022]
Abstract
PTPL1, a non-receptor type protein tyrosine phosphatase, has been involved in the regulation of apoptosis and invasiveness of various tumour cell types, but its role in prostate cancer remained to be investigated. We report here that downregulation of PTPL1 by small interfering RNA in PC3 cells decreases cell proliferation and concomitantly reduces the expression of cell cycle-related proteins such as cyclins E and B1, PCNA, PTTG1 and phospho-histone H3. PTPL1 downregulation also increases the invasion ability of PC3 cells through Matrigel coated membranes. cDNA array of PTPL1-silenced PC3 cells versus control cells showed an upregulation of invasion-related genes such as uPA, uPAR, tPA, PAI-1, integrin α6 and osteopontin. This increased expression was also confirmed in PTPL1-silenced DU145 prostate cancer cells by quantitative real time PCR and western blot. These findings suggest that PTPL1 is an important mediator of central cellular processes such as proliferation and invasion.
Collapse
Affiliation(s)
- Carolina Castilla
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avenida Manuel Siurot s/n, 41013 Seville, Spain
| | | | | | | | | | | | | |
Collapse
|
23
|
Schooley AM, Andrews NM, Zhao H, Addison CL. β1 integrin is required for anchorage-independent growth and invasion of tumor cells in a context dependent manner. Cancer Lett 2011; 316:157-67. [PMID: 22099877 DOI: 10.1016/j.canlet.2011.10.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 10/12/2011] [Accepted: 10/23/2011] [Indexed: 10/15/2022]
Abstract
Recent studies suggest that extracellular matrix (ECM) components within the tumor microenvironment can influence malignant progression, thus we investigated the influence of the ECM binding receptor β1 integrin, on the hallmark properties of tumorigenesis. Small interfering (si) or short hairpin (sh) RNA approaches were used to deplete β1 integrin in cancer cell lines. β1 integrin-depleted cells were then assessed for their growth and invasive capabilities using 2-dimensional (2D) or 3D culture conditions. Depletion of β1 integrin expression did not impact cell growth in 2D assay systems; however, β1 integrin and its ligand fibronectin were required for growth in 3D. β1 integrin-depleted cells also had reduced invasive capabilities, in part due to increased tissue inhibitor of metalloprotease (TIMP)-2 expression in conjunction with down-regulation of matrix metalloprotease (MMP)-9 levels in β1 integrin-depleted cells. Our results suggest that despite no apparent effect on 2D cell growth, fibronectin-β1 integrin signaling is a critical mediator of the 3D growth and invasive properties of tumor cells. These observations highlight the importance of investigating the role of adhesion molecules in the appropriate context and furthermore identify β1 integrin as a possible therapeutic target to inhibit the aggressive growth and invasion of tumor cells.
Collapse
Affiliation(s)
- Allana M Schooley
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| | | | | | | |
Collapse
|
24
|
Pontes-Junior J, Reis ST, Dall'Oglio M, Neves de Oliveira LC, Cury J, Carvalho PA, Ribeiro-Filho LA, Moreira Leite KR, Srougi M. Evaluation of the expression of integrins and cell adhesion molecules through tissue microarray in lymph node metastases of prostate cancer. J Carcinog 2011; 8:3. [PMID: 19240373 PMCID: PMC2678866 DOI: 10.4103/1477-3163.48453] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Integrins and adhesion molecules are responsible for the maintenance of the epithelial phenotype. Cell culture studies have reported the correlation between adhesion molecule expression and prostate carcinoma, but their role in the metastatic process is not yet known. Our aim is to study the expression profiles of these molecules and evaluate their association with the metastatic behavior of prostate adenocarcinoma. Materials and Methods: A Tissue Microarray containing two samples from 19 primary tumors and one from their corresponding lymph node metastases was constructed and subjected to immunohistochemical analysis of the expression of integrins, E-cadherin and β and γ-catenins. Within each case, paired analyses were also performed to evaluate gains or losses in metastasis compared to its primary tumor. Results: The expression of αv, αvβ3, α2β1 and γ-catenin were abnormal in almost every case. Marked loss of E-cadherin and β4 integrin was found in primary and metastatic lesions. β-catenin was normal in all primary cases and in 94% of metastases. α6 was normal in all primary tumors and metastases. α3 and α3β1 were normal in 32% of primary cases and in 53% and 6% of metastases, respectively. In paired analyses, loss of E-cadherin, β4, αv, α3β1 and αvβ3 was found in 65%, 71%, 59%, 53% and 47% of patients, respectively. Catenins and α2β1 showed maintenance of expression in most of the cases. Conclusions: In this preliminary study we have shown that the loss of cell adhesion molecules can be considered a characteristic of the metastatic phenotype in prostate cancer. Larger series should be evaluated in order to confirm our findings.
Collapse
Affiliation(s)
- Jose Pontes-Junior
- Laboratory of Medical Investigation - LIM 55, Urology Department, Medical School University of São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Metastasis Update: Human Prostate Carcinoma Invasion via Tubulogenesis. Prostate Cancer 2011; 2011:249290. [PMID: 21949592 PMCID: PMC3177701 DOI: 10.1155/2011/249290] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 04/25/2011] [Indexed: 01/08/2023] Open
Abstract
This paper proposes that human prostate carcinoma primarily invades as a cohesive cell collective through a mechanism similar to embryonic tubulogenesis, instead of the popular epithelial-mesenchymal transformation (EMT) model. Evidence supporting a tubulogenesis model is presented, along with suggestions for additional research. Additionally, observations documenting cell adhesion molecule changes in tissue and stromal components are reviewed, allowing for comparisons between the current branching morphogenesis models and the tubulogenesis model. Finally, the implications of this model on prevailing views of therapeutic and diagnostic strategies for aggressive prostatic disease are considered.
Collapse
|
26
|
Cheng I, Plummer SJ, Neslund-Dudas C, Klein EA, Casey G, Rybicki BA, Witte JS. Prostate cancer susceptibility variants confer increased risk of disease progression. Cancer Epidemiol Biomarkers Prev 2010; 19:2124-32. [PMID: 20651075 PMCID: PMC2950095 DOI: 10.1158/1055-9965.epi-10-0268] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Genome-wide association studies have identified numerous single nucleotide polymorphisms (SNP) associated with the risk of prostate cancer. Our objective was to determine whether these SNPs affect the progression of prostate cancer. METHODS We genotyped 26 SNPs previously associated with prostate cancer risk among 788 aggressive prostate cancer patients who were treated by radical prostatectomy or radiation therapy. Prostate cancer progression was defined as biochemical recurrence based on posttreatment prostate-specific antigen levels of >0.3 ng/mL for radical prostatectomy patients or a 2-ng/mL increase above the nadir for radiation therapy patients, initiation of hormone treatment, or metastases. We assessed the association between independent and combined SNPs and disease progression by Cox proportional hazards regression. RESULTS Five SNPs showed independent associations with prostate cancer progression (rs12621278, rs629242, rs9364554, rs4430796, and rs5945572) based on stepwise regression analysis. The strongest SNP was rs12621278 in the ITGA6 locus, which was associated with a 2.4-fold increased risk of progression (P = 0.0003). When considering the sum of risk alleles across these five SNPs, each additional allele was associated with a 29% increase in risk of progression (95% confidence interval, 1.12-1-47). CONCLUSIONS We found that five of the recently highlighted prostate cancer susceptibility loci also influence prostate cancer progression beyond the known clinicopathologic predictors. If confirmed, these genetic variants might help clarify which tumors are likely to progress and require more aggressive treatment in contrast to those that might not have substantial effects on morbidity or mortality. IMPACT Genetic susceptibility variants for prostate cancer development may also inform disease progression.
Collapse
Affiliation(s)
- Iona Cheng
- Epidemiology Program, Cancer Research Center of Hawai`i, University of Hawai`i, Honolulu, HI 96813, USA
| | - Sarah J. Plummer
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Christine Neslund-Dudas
- Department of Biostatistics and Research Epidemiology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Eric A. Klein
- Department of Urology, Cleveland Clinic Taussig Cancer Institute and Glickman Urological and Kidney Institute, Cleveland, OH 44915, USA
| | - Graham Casey
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Benjamin A. Rybicki
- Department of Biostatistics and Research Epidemiology, Henry Ford Health System, Detroit, MI 48202, USA
| | - John S. Witte
- Departments of Epidemiology & Biostatistics and Urology, Institute for Human Genetics, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158-9001, USA
| |
Collapse
|
27
|
Lin E, Wang Q, Swenson S, Jadvar H, Groshen S, Ye W, Markland FS, Pinski J. The disintegrin contortrostatin in combination with docetaxel is a potent inhibitor of prostate cancer in vitro and in vivo. Prostate 2010; 70:1359-70. [PMID: 20623636 DOI: 10.1002/pros.21173] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND There are few available treatments for hormone refractory prostate cancer. Through the inhibition of integrins, contortrostatin (CN) effects tumor cell growth directly as well as through the inhibition of angiogenesis. The effect of CN in combination with docetaxel on prostate cancer cell lines in vitro and in vivo is evaluated in the present study. METHODS FACS analysis of integrin expression, assessment of CN and docetaxel exposure on viability of plated cancer cells, and scratch test migration analysis were performed on PC-3 prostate cancer cells. CN and docetaxel inhibition of both PC-3 and CWR-22 prostate cancer cell lines were evaluated in a mouse xenograft bone model. Angiogenic activity in tumors were assessed using IHC with antibodies to CD31. RESULTS Cell culture experiments indicate that the combination of docetaxel and CN inhibits growth in an additive fashion. FACS analysis of PC-3 cells shows expression of alpha5beta1 and alphavbeta5 integrins, but little expression of the alphavbeta3. CN showed complete inhibition of PC-3 migration in cultures grown on matrigel plates. In mice xenograft bone models, CN with docetaxel showed increased inhibition of both PC-3 and CWR-22 derived tumors. Analysis of treated xenograft tumors showed significantly decreased expression of CD31 indicating suppression of angiogenesis.
Collapse
Affiliation(s)
- Edwin Lin
- Department of Internal Medicine, Norris Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, California 90089, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Pontes-Júnior J, Reis ST, de Oliveira LCN, Sant'anna AC, Dall'oglio MF, Antunes AA, Ribeiro-Filho LA, Carvalho PA, Cury J, Srougi M, Leite KRM. Association between integrin expression and prognosis in localized prostate cancer. Prostate 2010; 70:1189-95. [PMID: 20564421 DOI: 10.1002/pros.21153] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Integrins and other adhesion molecules are essential for maintaining the epithelial phenotype. Some studies have reported correlations between abnormalities in their expression and carcinogenesis, but their role in prostate cancer is unclear. Our aim was to study the expression profile of integrins in surgical specimens of prostate cancer and associate their expression patterns with patient outcomes. METHODS We selected 111 patients with localized prostate cancer who had undergone radical prostatectomy. Of these patients, 60 had no tumor recurrence after a median follow-up of 123 months. Integrin expression was evaluated by immunohistochemistry in a tissue microarray containing two tumor samples per patient. A semiquantitative analysis was employed. We measured the association between the expression of eight integrins and tumor recurrence. RESULTS Multivariate analysis showed that expression of alpha3 and alpha3beta1 was related to worse outcome. When alpha3 expression was strong and alpha3beta1 expression was positive, the odds of recurrence were 3.0- and 2.5-fold higher, respectively. Only 19% and 28% of patients were recurrence-free in a mean period of 123 months of follow up when their tumors showed strong alpha3 or positive alpha3beta1 immuno-expression, respectively. CONCLUSIONS We have shown that the expression of integrin alpha3beta1 was independently associated with tumor recurrence after radical prostatectomy, suggesting that this integrin is a potential prognostic marker.
Collapse
Affiliation(s)
- José Pontes-Júnior
- Laboratory of Medical Investigation (LIM55), Urology Department, University of Sao Paulo Medical School, Sao Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Khamis ZI, Iczkowski KA, Sahab ZJ, Sang QXA. Protein profiling of isolated leukocytes, myofibroblasts, epithelial, Basal, and endothelial cells from normal, hyperplastic, cancerous, and inflammatory human prostate tissues. J Cancer 2010; 1:70-9. [PMID: 20842227 PMCID: PMC2938068 DOI: 10.7150/jca.1.70] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
In situ neoplastic prostate cells are not lethal unless they become invasive and metastatic. For cells to become invasive, the prostate gland must undergo degradation of the basement membrane and disruption of the basal cell layer underneath the luminal epithelia. Although the roles of proteinases in breaking down the basement membrane have been well-studied, little is known about the factors that induce basal cell layer disruption, degeneration, and its eventual disappearance in invasive cancer. It is hypothesized that microenvironmental factors may affect the degradation of the basal cell layer, which if protected may prevent tumor progression and invasion. In this study, we have revealed differential protein expression patterns between epithelial and stromal cells isolated from different prostate pathologies and identified several important epithelial and stromal proteins that may contribute to inflammation and malignant transformation of human benign prostate tissues to cancerous tissues using matrix-assisted laser desorption ionization time-of-flight mass spectrometry and proteomics methods. Cellular retinoic acid-binding protein 2 was downregulated in basal cells of benign prostate. Caspase-1 and interleukin-18 receptor 1 were highly expressed in leukocytes of prostate cancer. Proto-oncogene Wnt-3 was downregulated in endothelial cells of prostatitis tissue and tyrosine phosphatase non receptor type 1 was only found in normal and benign endothelial cells. Poly ADP-ribose polymerase 14 was downregulated in myofibroblasts of prostatitis tissue. Interestingly, integrin alpha-6 was upregulated in epithelial cells but not detected in myofibroblasts of prostate cancer. Further validation of these proteins may generate new strategies for the prevention of basal cell layer disruption and subsequent cancer invasion.
Collapse
Affiliation(s)
- Zahraa I. Khamis
- 1. Department of Chemistry and Biochemistry and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Kenneth A. Iczkowski
- 2. Department of Pathology, University of Colorado Health Science Center, Aurora, CO 80045, USA
| | - Ziad J. Sahab
- 1. Department of Chemistry and Biochemistry and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Qing-Xiang Amy Sang
- 1. Department of Chemistry and Biochemistry and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
30
|
Kielosto M, Nummela P, Järvinen K, Yin M, Hölttä E. Identification of integrins alpha6 and beta7 as c-Jun- and transformation-relevant genes in highly invasive fibrosarcoma cells. Int J Cancer 2009; 125:1065-73. [PMID: 19405119 DOI: 10.1002/ijc.24391] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Understanding the mechanisms of tumor cell invasion is essential for our attempts to prevent cancer deaths. We screened by DNAmicroarrays the c-Jun- and transformation-related gene expression changes in S-adenosylmethionine decarboxylase (AdoMetDC)-overexpressing mouse fibroblasts that are highly invasive in vivo, and their derivatives expressing a tetracycline-inducible dominant-negative mutant of c-Jun (TAM67) or c-Jun shRNA. Among the small set of target genes detected were integrins alpha6 and beta7, cathepsin L and thymosin beta4, all upregulated in the AdoMetDC-transformed cells and downregulated upon reversal of transformation by TAM67 or c-Jun shRNA. The upregulation of integrin alpha6 subunit, pairing with integrin beta1, endowed the transformed cells with the capability to attach to basement membrane laminin and to spread. Further, inhibition of integrin alpha6 or beta1 function with neutralizing antibodies blocked the invasiveness of AdoMetDC-transformants and human HT-1080 fibrosarcoma cells in three-dimensional Matrigel. Moreover, immunohistochemical analyses showed strong integrin alpha6 staining in high-grade human fibrosarcomas. Our data show that c-Jun can regulate all three key steps of invasion: cell adhesion (integrin alpha6), basement membrane/extracellular matrix degradation (cathepsin L) and cell migration (thymosin beta4). In addition, this is the first study to associate integrin beta7, known as a leukocyte-specific integrin binding to endothelial/epithelial cell adhesion molecules, with the transformed phenotype in cells of nonleukocyte origin. As tumor cell invasion is a prerequisite for metastasis, the observed critical role of integrin alpha6beta1 in fibrosarcoma cell invasion/spreading allures testing antagonists to integrin alpha6beta1, alone or combined with inhibitors of cathepsin L and thymosin beta4, as chemotherapeutic agents.
Collapse
Affiliation(s)
- Mari Kielosto
- Haartman Institute, Department of Pathology, University of Helsinki and Helsinki University Central Hospital, Finland
| | | | | | | | | |
Collapse
|
31
|
Delamarre E, Taboubi S, Mathieu S, Bérenguer C, Rigot V, Lissitzky JC, Figarella-Branger D, Ouafik L, Luis J. Expression of integrin alpha6beta1 enhances tumorigenesis in glioma cells. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:844-55. [PMID: 19574430 DOI: 10.2353/ajpath.2009.080920] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The integrin alpha6beta1 and its main ligand laminin-111 are overexpressed in glioblastoma, as compared with normal brain tissue, suggesting they may be involved in glioblastoma malignancy. To address this question, we stably expressed the alpha6 integrin subunit in the U87 cell line via retroviral-mediated gene transfer. We show that cell surface expression of the alpha6beta1 integrin led to dramatic changes in tumor U87 cell behavior, both in vitro and in vivo. Nude mice receiving either subcutaneous or intracerebral inoculation of alpha6beta1-expressing cells developed substantially more voluminous tumors than mice injected with control cells. The difference in tumor growth was associated with a marked increase in vascularization in response to alpha6beta1 integrin expression and may also be related to changes in the balance between cell proliferation and survival. Indeed, expression of alpha6beta1 enhanced proliferation and decreased apoptosis of U87 cells both in the tumor and in vitro. Additionally, we demonstrate that alpha6beta1 is implicated in glioblastoma cell migration and invasion and that laminin-111 might mediate dissemination of alpha6beta1-positive cells in vivo. Our results highlight for the first time the considerable role of the integrin alpha6beta1 in glioma progression.
Collapse
Affiliation(s)
- Estelle Delamarre
- INSERM U911 (CRO2), Faculté de Pharmacie, 27, Bd J. Moulin, 13 385 Marseille Cedex 5, France
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Zhang X, Fournier MV, Ware JL, Bissell MJ, Jacoub A, Zehner ZE. Inhibition of vimentin or beta1 integrin reverts morphology of prostate tumor cells grown in laminin-rich extracellular matrix gels and reduces tumor growth in vivo. Mol Cancer Ther 2009; 8:499-508. [PMID: 19276168 PMCID: PMC2703491 DOI: 10.1158/1535-7163.mct-08-0544] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Prostate epithelial cells grown embedded in laminin-rich extracellular matrix (lrECM) undergo morphologic changes that closely resemble their architecture in vivo. In this study, growth characteristics of three human prostate epithelial sublines derived from the same cellular lineage, but displaying different tumorigenic and metastatic properties in vivo, were assessed in three-dimensional lrECM gels. M12, a highly tumorigenic and metastatic subline, was derived from the immortalized, prostate epithelial P69 cell line by selection in athymic, nude mice and found to contain a deletion of 19p-q13.1. The stable reintroduction of an intact human chromosome 19 into M12 resulted in a poorly tumorigenic subline, designated F6. When embedded in lrECM gels, the parental, nontumorigenic P69 line produced acini with clearly defined lumena. Immunostaining with antibodies to beta-catenin, E-cadherin, or alpha6 and beta1 integrins showed polarization typical of glandular epithelium. In contrast, the metastatic M12 subline produced highly disorganized cells with no evidence of polarization. The F6 subline reverted to acini-like structures exhibiting basal polarity marked with integrins. Reducing either vimentin levels via small interfering RNA interference or the expression of alpha6 and beta1integrins by the addition of blocking antibodies, reorganized the M12 subline into forming polarized acini. The loss of vimentin significantly reduced M12-Vim tumor growth when assessed by s.c. injection in athymic mice. Thus, tumorigenicity in vivo correlated with disorganized growth in three-dimensional lrECM gels. These studies suggest that the levels of vimentin and beta1 integrin play a key role in the homeostasis of the normal acinus in prostate and that their dysregulation may lead to tumorigenesis.
Collapse
Affiliation(s)
- Xueping Zhang
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University-Medical Campus, Richmond VA 23298
| | - Marcia V. Fournier
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Joy L. Ware
- Department of Pathology, Massey Cancer Center, Virginia Commonwealth University-Medical Campus, Richmond VA 23298
| | - Mina J. Bissell
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Adly Jacoub
- Department of Radiation Oncology, Massey Cancer Center, Virginia Commonwealth University-Medical Campus, Richmond VA 23298
| | - Zendra E. Zehner
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University-Medical Campus, Richmond VA 23298
| |
Collapse
|
33
|
Clarke NW, Hart CA, Brown MD. Molecular mechanisms of metastasis in prostate cancer. Asian J Androl 2009; 11:57-67. [PMID: 19050684 PMCID: PMC3735202 DOI: 10.1038/aja.2008.29] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Accepted: 10/11/2008] [Indexed: 01/30/2023] Open
Abstract
Prostate cancer (PCa) preferentially metastasizes to the bone marrow stroma of the axial skeleton. This activity is the principal cause of PCa morbidity and mortality. The exact mechanism of PCa metastasis is currently unknown, although considerable progress has been made in determining the key players in this process. In this review, we present the current understanding of the molecular processes driving PCa metastasis to the bone.
Collapse
Affiliation(s)
- Noel W. Clarke
- Genito-Urinary Cancer Research Group, School of Cancer and Imaging Sciences, Paterson Institute for Cancer Research, Christie Hospital, University of Manchester, Manchester M20 4BX, UK
| | - Claire A. Hart
- Genito-Urinary Cancer Research Group, School of Cancer and Imaging Sciences, Paterson Institute for Cancer Research, Christie Hospital, University of Manchester, Manchester M20 4BX, UK
| | - Mick D. Brown
- Genito-Urinary Cancer Research Group, School of Cancer and Imaging Sciences, Paterson Institute for Cancer Research, Christie Hospital, University of Manchester, Manchester M20 4BX, UK
| |
Collapse
|
34
|
King TE, Pawar SC, Majuta L, Sroka IC, Wynn D, Demetriou MC, Nagle RB, Porreca F, Cress AE. The role of alpha 6 integrin in prostate cancer migration and bone pain in a novel xenograft model. PLoS One 2008; 3:e3535. [PMID: 18958175 PMCID: PMC2570216 DOI: 10.1371/journal.pone.0003535] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2008] [Accepted: 09/26/2008] [Indexed: 01/13/2023] Open
Abstract
Of the estimated 565,650 people in the U.S. who will die of cancer in 2008, almost all will have metastasis. Breast, prostate, kidney, thyroid and lung cancers metastasize to the bone. Tumor cells reside within the bone using integrin type cell adhesion receptors and elicit incapacitating bone pain and fractures. In particular, metastatic human prostate tumors express and cleave the integrin A6, a receptor for extracellular matrix components of the bone, i.e., laminin 332 and laminin 511. More than 50% of all prostate cancer patients develop severe bone pain during their remaining lifetime. One major goal is to prevent or delay cancer induced bone pain. We used a novel xenograft mouse model to directly determine if bone pain could be prevented by blocking the known cleavage of the A6 integrin adhesion receptor. Human tumor cells expressing either the wildtype or mutated A6 integrin were placed within the living bone matrix and 21 days later, integrin expression was confirmed by RT-PCR, radiographs were collected and behavioral measurements of spontaneous and evoked pain performed. All animals independent of integrin status had indistinguishable tumor burden and developed bone loss 21 days after surgery. A comparison of animals containing the wild type or mutated integrin revealed that tumor cells expressing the mutated integrin resulted in a dramatic decrease in bone loss, unicortical or bicortical fractures and a decrease in the ability of tumor cells to reach the epiphyseal plate of the bone. Further, tumor cells within the bone expressing the integrin mutation prevented cancer induced spontaneous flinching, tactile allodynia, and movement evoked pain. Preventing A6 integrin cleavage on the prostate tumor cell surface decreased the migration of tumor cells within the bone and the onset and degree of bone pain and fractures. These results suggest that strategies for blocking the cleavage of the adhesion receptors on the tumor cell surface can significantly prevent cancer induced bone pain and slow disease progression within the bone. Since integrin cleavage is mediated by Urokinase-type Plasminogen Activator (uPA), further work is warranted to test the efficacy of uPA inhibitors for prevention or delay of cancer induced bone pain.
Collapse
Affiliation(s)
- Tamara E. King
- Department of Pharmacology, The Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
- Department of Pathology, The Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
| | - Sangita C. Pawar
- Department of Cell Biology & Anatomy, The Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
- Department of Pathology, The Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
| | - Lisa Majuta
- Department of Pharmacology, The Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
| | - Isis C. Sroka
- Department of Cell Biology & Anatomy, The Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
| | - Danyel Wynn
- Department of Physiology, The Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
| | | | - Raymond B. Nagle
- Department of Pathology, The Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
| | - Frank Porreca
- Department of Pharmacology, The Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
| | - Anne E. Cress
- Department of Cell Biology & Anatomy, The Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
35
|
Sroka IC, Chen ML, Cress AE. Simplified purification procedure of laminin-332 and laminin-511 from human cell lines. Biochem Biophys Res Commun 2008; 375:410-3. [PMID: 18713621 PMCID: PMC2573946 DOI: 10.1016/j.bbrc.2008.08.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Accepted: 08/05/2008] [Indexed: 10/21/2022]
Abstract
Laminins are glycoproteins expressed in the basement membrane of multiple epithelial tissues. Previously described purification procedures for the human laminin variants laminin-5 (LN-332) and laminin-10 (LN-511) use tissue as starting material and have multiple steps. We demonstrate a two-step laminin immunoaffinity purification method to produce consistent quantities of intact and biologically active LN-332 and LN-511 from human keratinocyte (HaCaT) and human lung carcinoma (A549) cell lines, respectively. The purification of LN-332 and LN-551 was demonstrated by PAGE analysis, silver staining and Western blot analysis. The purification procedure includes instruction on removing a cell adhesion contaminant known as galectin-3 binding protein from purified LN-511. The biological activity of purified laminin was tested in a standard cell adhesion assay and compared to commercially available LN-111. This rapid and reproducible purification method will contribute to understanding the role of LN-332 and LN-511 in cell behavior, signaling, and gene expression.
Collapse
Affiliation(s)
- Isis C. Sroka
- Department of Cell Biology and Anatomy, University of Arizona, Tucson, Arizona
| | - Man Ling Chen
- Department of Cell Biology and Anatomy, University of Arizona, Tucson, Arizona
| | - Anne E. Cress
- Department of Cell Biology and Anatomy, University of Arizona, Tucson, Arizona
- The Arizona Cancer Center, University of Arizona, Tucson, Arizona 85724
| |
Collapse
|
36
|
Mook ORF, van Marle J, Jonges R, Vreeling-Sindelárová H, Frederiks WM, Van Noorden CJF. Interactions between colon cancer cells and hepatocytes in rats in relation to metastasis. J Cell Mol Med 2008; 12:2052-61. [PMID: 18208563 PMCID: PMC4506170 DOI: 10.1111/j.1582-4934.2008.00242.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Adhesion of cancer cells to endothelium is considered an essential step in metastasis. However, we have shown in a previous study that when rat colon cancer cells are administered to the vena portae, they get stuck mechanically in liver sinusoids. Then, endothelial cells retract rapidly and cancer cells bind to hepatocytes. We investigated the molecular nature of these interactions between colon cancer cells and hepatocytes. Cancer cells in coculture with hepatocytes became rapidly activated with distinct morphological changes. Cancer cells formed long cytoplasmic protrusions towards hepatocytes in their close vicinity and these protrusions attached to microvilli of hepatocytes. Then, adhering membrane areas were formed by both cell types. Integrin subunits alphav, alpha6 and beta1 but not alphaL, beta2, beta3 and CD44 and CD44v6 were expressed on the cancer cells. In conclusion, colon cancer cells show an active behaviour to bind to hepatocytes, likely involving the integrin subunits alphav, alpha6 and beta1, indicating that early events in colon cancer metastasis in liver are distinctly different than assumed thus far.
Collapse
Affiliation(s)
- O R F Mook
- Department of Cell Biology and Histology, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
37
|
Thomas S, Chiriva-Internati M, Shah GV. Calcitonin receptor-stimulated migration of prostate cancer cells is mediated by urokinase receptor-integrin signaling. Clin Exp Metastasis 2007; 24:363-77. [PMID: 17487556 DOI: 10.1007/s10585-007-9073-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2007] [Accepted: 03/31/2007] [Indexed: 12/25/2022]
Abstract
Abundance of calcitonin (CT) and calcitonin receptor (CTR) mRNA in primary prostate tumors positively correlates with tumor grade, and exogenously added CT increases the invasion of prostate cancer cell lines. We examined acute and chronic actions of CT on migration of highly metastatic PC-3M cells and poorly invasive LNCaP cells on several extracellular matrices in a spheroid disaggregation/migration assay. While PC-3M spheroids displayed maximum disaggregation/migration on vitronectin (VN), LNCaP spheroids preferred collagen but also migrated significantly on VN. Up-regulation of CT significantly enhanced disaggregation/migration of PC-3M spheroids on VN, but not on fibronectin. In contrast, down-regulation of CT, CTR, protein kinase A or urokinase-type plasminogen activator receptor (uPAR) led to amelioration of PC-3M spheroid disaggregation/migration. CT selectively increased surface activity of alpha v beta 3 or alpha 6 beta 5 integrins in PC-3M and LNCaP cell lines, respectively, and uPAR-integrin association. Finally, either CT or urokinase could completely restore migration of CT-knock-down PC-3M spheroids. But, only forced expression of urokinase receptor coupled with exogenous addition of urokinase restored migration of CTR-knock-down spheroids. These results support our hypothesis that up-regulation of CT biosynthesis and activation of CT-CTR axis in primary prostate tumors may have direct relevance in their progression to the metastatic phenotype.
Collapse
Affiliation(s)
- Shibu Thomas
- Department of Pharmacology, University of Louisiana College of Pharmacy, Monroe, LA 71209, USA
| | | | | |
Collapse
|
38
|
Stevenson M, Hale ABH, Hale SJ, Green NK, Black G, Fisher KD, Ulbrich K, Fabra A, Seymour LW. Incorporation of a laminin-derived peptide (SIKVAV) on polymer-modified adenovirus permits tumor-specific targeting via alpha6-integrins. Cancer Gene Ther 2007; 14:335-45. [PMID: 17235355 DOI: 10.1038/sj.cgt.7701022] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2006] [Revised: 09/30/2006] [Accepted: 11/18/2006] [Indexed: 01/17/2023]
Abstract
Effective gene therapy for disseminated metastatic cancer is currently impossible because of poor delivery of vector to target sites. Modification of viral vectors to target advanced cancer has long been a challenge. In this study, we aimed to redirect adenovirus tropism to infect prostate cancer cells via alpha6beta1 integrins, whose expression is upregulated during prostate cancer progression. To ablate normal mechanisms of infection and provide a framework for attachment of targeting ligands, viruses were non-genetically modified with pHPMA-ONp polymer. Addition of polymer-coated virus to prostate cells showed significantly reduced transgene expression compared with unmodified virus. To restore infectivity, an alpha6-integrin binding peptide (-SIKVAV-) derived from laminin was incorporated onto the surface of the polymer-coated viruses. Photon correlation spectroscopic analysis revealed a small increase in the mean diameter of the particles following retargeting. Addition of -SIKVAV- peptide restored virus infectivity of PC-3 cells in a ligand concentration-dependent manner that was significantly improved following removal of unincorporated polymer and peptide. Competition assays using cells preincubated with Ad5 fiber protein or free -SIKVAV- peptide confirmed that entry of retargeted viruses was mediated via the incorporated ligand. Application of retargeted viruses to a panel of human cell lines revealed varying levels of transduction efficiency. Flow cytometric analysis of cells using anti-alpha6 integrin and anti-beta1 integrin antibodies demonstrated that for prostate cells, greater transduction efficiency correlated with higher levels of expression of both integrin subunits. Furthermore with the exception of LNCaP cells, increased alpha6beta1 integrin expression correlated with advanced disease. Intravenous administration of retargeted viruses to tumor-bearing mice resulted in slower plasma clearance and greatly reduced liver tropism, and hence toxicity compared with unmodified virus, while maintaining reporter gene expression in the tumor. The data suggest that YESIKVAVS-retargeted viruses have potential for systemic delivery for the treatment of metastatic disease.
Collapse
Affiliation(s)
- M Stevenson
- Department of Clinical Pharmacology, University of Oxford, Oxford, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Malik M, Gürcan HM, Christen W, Ahmed AR. Relationship between cancer and oral pemphigoid patients with antibodies to α6-integrin. J Oral Pathol Med 2006; 36:1-5. [PMID: 17181734 DOI: 10.1111/j.1600-0714.2006.00483.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Mucous membrane pemphigoid is an autoimmune mucocutaneous blistering disease. A subset, known as anti-epiligrin cicatricial pemphigoid is associated with a high risk for malignancy. Oral pemphigoid (OP) is limited to the oral cavity. The purpose of this study was to determine the association between malignancy and patients with OP with antibodies to alpha6-integrin subunit. METHODS We determined the incidence of cancer in 72 patients with OP and compared it to the expected incidence using age and sex-specific rates of malignancy in the National Cancer Institute's Surveillance, Epidemiology, and End Results (NCI SEER) Registry. RESULTS During a mean observation period of 9.1 years (range: 2.8-40), for 70, three OP patients developed malignancies. The expected number of cancers based on the NCI SEER Registry was 8.83. The relative risk for cancer in OP patients, with autoantibodies to alpha6-integrin, was 0.34 (95% CI, 0.07-0.99, P < 0.05). CONCLUSION It appears that patients with OP, with antibodies to alpha6, may have a possible reduced relative risk for developing cancer.
Collapse
Affiliation(s)
- Mohsin Malik
- Department of Medicine, New England Baptist Hospital, Harvard School of Dental Medicine, Boston, MA, USA
| | | | | | | |
Collapse
|
40
|
Knudsen BS, Miranti CK. The impact of cell adhesion changes on proliferation and survival during prostate cancer development and progression. J Cell Biochem 2006; 99:345-61. [PMID: 16676354 DOI: 10.1002/jcb.20934] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In the normal prostate epithelium, androgen receptor (AR) negative basal epithelial cells adhere to the substratum, while AR expressing secretory cells lose substratum adhesion. In contrast, prostate cancer cells both express AR and adhere to a tumor basement membrane. In this review, we describe the differential expression of integrins, growth factor receptors (GFRs), and AR in normal and cancerous epithelium. In addition, we discuss how signals from integrins, GFRs, and AR are integrated to regulate the proliferation and survival of normal and malignant prostate epithelial cells. While cell adhesion is likely of great importance when considering therapeutic approaches for treatment of metastatic prostate cancer, no data on integrin expression are available from tissues of prostate cancer metastasis. However, several drug targets that are upregulated after androgen ablative therapy regulate cell adhesion and thus novel targeted therapies indirectly interfere with cell adhesion mechanisms in prostate cancer cells.
Collapse
Affiliation(s)
- Beatrice S Knudsen
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA.
| | | |
Collapse
|
41
|
Sroka TC, Pennington ME, Cress AE. Synthetic D-amino acid peptide inhibits tumor cell motility on laminin-5. Carcinogenesis 2006; 27:1748-57. [PMID: 16537560 PMCID: PMC4069207 DOI: 10.1093/carcin/bgl005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cell motility is partially dependent on interactions between the integrins and the extracellular matrix. Our previous studies have identified synthetic D-amino acid cell adhesion peptides using a combinatorial screening approach. In this study, we demonstrate that HYD1 (kikmviswkg) completely blocks random haptotactic migration and inhibits invasion of prostate carcinoma cells on laminin-5. This effect is adhesion independent and reversible. The inhibition of migration by HYD1 involves a dramatic remodeling of the actin cytoskeleton resulting in increased stress fiber formation and actin colocalization with cortactin at the cell membrane. HYD1 interacts with alpha6beta1 (not alpha6beta4) and alpha3beta1 integrins and surprisingly elevates laminin-5-dependent intracellular signals including focal adhesion kinase, mitogen-activated protein kinase kinase and extracellular signal-regulated kinase. HYD1 does not contain a previously characterized binding sequence for integrins. A scrambled derivative of HYD1, called HYDS (wiksmkivkg), does not interact with the alpha6 or alpha3 integrin subunits and is not biologically active. Taken together, these results indicate that HYD1 is a biologically active integrin-targeting peptide that reversibly inhibits tumor cell migration on laminin-5 and uncouples phosphotyrosine signaling from cytoskeletal-dependent migration.
Collapse
Affiliation(s)
- Thomas C. Sroka
- Department of Cell Biology and Anatomy, The University of Arizona, Tucson, AZ 85724, USA
| | - Michael E. Pennington
- Department of Cell Biology and Anatomy, The University of Arizona, Tucson, AZ 85724, USA
| | - Anne E. Cress
- Department of Cell Biology and Anatomy, The University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
42
|
Chu JH, Sun ZY, Meng XL, Wu JH, He GL, Liu GM, Jiang XR. Differential metastasis-associated gene analysis of prostate carcinoma cells derived from primary tumor and spontaneous lymphatic metastasis in nude mice with orthotopic implantation of PC-3M cells. Cancer Lett 2006; 233:79-88. [PMID: 15885894 DOI: 10.1016/j.canlet.2005.03.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2005] [Revised: 02/20/2005] [Accepted: 03/01/2005] [Indexed: 01/29/2023]
Abstract
The purpose of these studies was to explore the genes associated with invasion and metastasis of human prostatic carcinoma line PC-3M in nude mice. After PC-3M cells were inoculated in orthotopic site (prostate) in male nude mice for two months, tumor cells were isolated from primary tumor and lymph node metastasis in the same mouse, respectively. Cell invasion and adhesion ability in vitro were first compared between two cell lines. Then human metastasis-related genes differentially expressed between them were analyzed by utilizing cDNA microarray technique. The in vitro cell invasion and adhesion potential of tumor cells from lymph node metastasis was significantly higher than those from primary tumor, Metastasis-related genes differentially expressed between those two cell lines were identified, all of them were up-regulated in the tumor cells from lymph node metastasis and could be categorized as: (1) genes encoding cellular matrix-degrading proteolytic enzyme including cathepsin and MMP; (2) genes encoding transcription factors; (3) genes related to heterotypic adhesion of tumor cells; (4) genes encoding cell surface receptors. Moreover, Four genes were chosen for semi-quantitative RT-PCR analysis, they showed a consistent expression pattern with that of cDNA microarray analysis. We concluded that the lymph node metastasis in nude mice given an injection of PC-3M cells in the prostate is a selective process favoring the survival and growth of a special subpopulation derived from primary tumor with specific genetic alterations, which may play a pivotal role in the metastasis of prostate cancer. Identification and further characterization of these genes may allow a better understanding of lymphatic metastasis in prostate carcinoma.
Collapse
Affiliation(s)
- Jian Hong Chu
- Department of Pharmacology and Toxicology, Shanghai Institute of Planned Parenthood Research, Fudan University. National Evaluation Center for the Toxicology of Fertility and Regulating Drugs, Shanghai, 200032, P.R. China
| | | | | | | | | | | | | |
Collapse
|
43
|
Sawai H, Okada Y, Funahashi H, Matsuo Y, Takahashi H, Takeyama H, Manabe T. Interleukin-1alpha enhances the aggressive behavior of pancreatic cancer cells by regulating the alpha6beta1-integrin and urokinase plasminogen activator receptor expression. BMC Cell Biol 2006; 7:8. [PMID: 16504015 PMCID: PMC1388210 DOI: 10.1186/1471-2121-7-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2005] [Accepted: 02/20/2006] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND In human pancreatic cancer progression, the alpha6beta1-integrin is expressed on cancer cell surface during invasion and metastasis formation. In this study, we investigated whether interleukin (IL)-1alpha induces the alterations of integrin subunits and urokinase plasminogen activator/urokinase plasminogen activator receptor (uPA/uPAR) expression in pancreatic cancer cells. We hypothesize that the alterations of integrin subunits and uPA/uPAR expression make an important role in signaling pathways responsible for biological behavior of pancreatic cancer cells. RESULTS IL-1alpha upregulated the expression of alpha6 and beta1 integrins without any alterations of alpha5 and alphav integrins expression. IL-1alpha also induced enhancement in the expression of uPA/uPAR in pancreatic cancer cells. IL-1alpha enhanced the proliferation, adhesion, and migration in pancreatic cancer cells, and IL-1alpha-induced alterations of uPA/uPAR expression correlated with the increased the migration of pancreatic cancer cells. Upregulation of alpha6 integrin subunit and uPA/uPAR correlated with the activation of Ras and downstream extracellular signal-regulated kinase (ERK) pathways. IL-1alpha-induced activation of Ras and downstream ERK can be inhibited by using inhibitory antibodies against alpha6 and beta1 integrin and uPAR, consistent with the inhibition of proliferation, adhesion and migration of pancreatic cancer cells. Immunohistochemical analysis demonstrated a significant association between strong expressions of alpha6 integrin with uPAR in pancreatic cancer specimens. Furthermore, the strong expression of alpha6 integrin and uPAR was found to be independent prognosticator in pancreatic cancer patients. CONCLUSION Based on these findings, we conclude that IL-1alpha can induce selective upregulation of alpha6beta1-integrin and uPA/uPAR in pancreatic cancer cells and these changes may modulate the aggressive functions of pancreatic cancer.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Carcinoma, Pancreatic Ductal/chemistry
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/pathology
- Cell Adhesion/drug effects
- Cell Division/drug effects
- Cell Line, Tumor/drug effects
- Cell Line, Tumor/metabolism
- Cell Line, Tumor/pathology
- Cell Movement/drug effects
- Extracellular Signal-Regulated MAP Kinases/physiology
- Female
- Humans
- Integrin alpha5/analysis
- Integrin alpha6beta1/biosynthesis
- Integrin alpha6beta1/genetics
- Integrin alphaV/analysis
- Integrin beta4/analysis
- Interleukin-1/pharmacology
- Laminin/metabolism
- Life Tables
- Male
- Middle Aged
- Neoplasm Invasiveness/physiopathology
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Pancreatic Neoplasms/chemistry
- Pancreatic Neoplasms/mortality
- Pancreatic Neoplasms/pathology
- Proto-Oncogene Proteins p21(ras)/physiology
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/genetics
- Receptors, Urokinase Plasminogen Activator
- Recombinant Fusion Proteins/pharmacology
- Signal Transduction/drug effects
- Survival Analysis
- Up-Regulation/drug effects
- Urokinase-Type Plasminogen Activator/biosynthesis
- Urokinase-Type Plasminogen Activator/genetics
Collapse
Affiliation(s)
- Hirozumi Sawai
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Yuji Okada
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Hitoshi Funahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Hiromitsu Takeyama
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Tadao Manabe
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| |
Collapse
|
44
|
Ayala GE, Dai H, Li R, Ittmann M, Thompson TC, Rowley D, Wheeler TM. Bystin in perineural invasion of prostate cancer. Prostate 2006; 66:266-72. [PMID: 16245277 DOI: 10.1002/pros.20323] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Bystin, Trophinin, and Tastin are component proteins of an adhesion molecule complex that plays a crucial role in the initial attachment of the embryo to the uterus. METHODS Profiling of genes differentially expressed in the perineural invasion (PNI) in vitro model by gene microarray analysis showed overexpression of bystin in prostate cancer cells co-cultured with nerves. Validation was performed at the RNA levels using quantitative PCR. RESULTS Bystin is overexpressed in cells co-cultured with nerves. Bystin is also present in human prostatic carcinoma (PCa) cells in PNI location in increasing gradient. Bystin is present in the supernatant of the PNI co-culture. CONCLUSIONS Their adhesive and invasive functions in the trophoblast suggest that they might also play a role in perineural adhesion. Bystin is, therefore, an important therapeutic target for neurotropic cancers.
Collapse
Affiliation(s)
- Gustavo E Ayala
- Department of Pathology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The progressive loss of laminin 5 and the alpha6beta4 integrin is a characteristic of the transition of prostatic intraepithelial neoplasia (PIN) to invasive human prostate cancer. Our objective was to determine if the loss of the interaction with laminin 5 would influence the ability of human epithelial cells to respond to DNA damage. Three cellular damage responses to ionizing radiation (IR) were analyzed including G2 progression, cdc2 phosphorylation, and cell survival. The adhesion of normal human prostate epithelial cells to laminin 5 amplified the G2 arrest induced by IR, and depends on a known cell binding domain of laminin 5. The alteration of G2 arrest was confirmed by an inhibition of phospho-cdc2 nuclear translocation. In contrast, a prostate epithelial cancer cell line blocked in G2 independent of adhesion to laminin 5. The survival of these cell lines in response to IR was unaffected by adhesion to laminin 5. These results suggest that cell adhesion to laminin 5 in normal cells will amplify the IR induced G2 cell cycle progression block without altering cell survival. The loss of laminin 5 and the alpha6beta4 integrin in PIN lesions may contribute to the selection and progression of genetically unstable cell types via attenuation of a DNA damage induced G2 arrest.
Collapse
Affiliation(s)
| | - Monika Schmelz
- Southern Arizona Veterans Affair Health Care System, Tucson, Arizona
| | - Anne E. Cress
- Arizona Cancer Center, University of Arizona, Tucson, Arizona
- Department of Cell Biology and Anatomy, University of Arizona, Tucson, Arizona
- Correspondence to: Anne E. Cress, Arizona Cancer Center, 1515 N Campbell Ave, Tucson Arizona 85724. E-mail:
| |
Collapse
|
46
|
Zigrino P, Löffek S, Mauch C. Tumor-stroma interactions: their role in the control of tumor cell invasion. Biochimie 2005; 87:321-8. [PMID: 15781319 DOI: 10.1016/j.biochi.2004.10.025] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2004] [Accepted: 10/26/2004] [Indexed: 11/17/2022]
Abstract
The development and progression of tumors result from the concerted activity not only of tumor cells with neighboring cells e.g., fibroblasts and inflammatory cells. Host-tumor interactions are considered critical in tumor invasion and metastasis. In vitro studies as well as established in vivo models have analysed the reciprocal effects of tumor-host interactions for the tumor invasion process. These studies have shown that modifications in the extracellular matrix composition surrounding the tumors as well as alterations in the expression of tumor cell receptors or in the expression of growth factors/cytokines and proteases, are critical regulators of a developing tumor. We shortly review the most important and well characterized mechanisms involved in the progression of tumor cells through tissues, especially those participating in cellular communication, cell adhesion, and proteolysis.
Collapse
Affiliation(s)
- P Zigrino
- Department of Dermatology and Center for Molecular Medicine (CMMC), University of Cologne, Joseph-Stelzmann strasse, 50931 Cologne, Germany
| | | | | |
Collapse
|
47
|
Shen X, Falzon M. PTH-related protein enhances LoVo colon cancer cell proliferation, adhesion, and integrin expression. ACTA ACUST UNITED AC 2005; 125:17-27. [PMID: 15582709 DOI: 10.1016/j.regpep.2004.07.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2004] [Accepted: 07/21/2004] [Indexed: 12/27/2022]
Abstract
Parathyroid hormone-related protein (PTHrP) has been localized in human colon cancer tissue and cell lines. Tumor cell adhesion to extracellular matrix (ECM) proteins plays a major role in the invasion and metastasis of tumor cells, and is mediated via integrin subunits. The LoVo human colon cancer cell line was used as a model system to study the effects of PTHrP on cell proliferation and adhesion to ECM proteins found in normal liver. Clones of LoVo cells engineered to overexpress PTHrP by stable transfection with a PTHrP cDNA showed enhanced cell proliferation vs. control (empty vector-transfected) cells. PTHrP-overexpressing cells also showed significantly higher adhesion to collagen type I, fibronectin, and laminin, and enhanced expression of the [symbol: see text] integrin subunits. These results indicate that PTHrP may play a role in colon cancer invasion and metastasis by increasing cell proliferation and adhesion to the ECM via upregulation of proinvasive integrin expression.
Collapse
Affiliation(s)
- Xiaoli Shen
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 10th and Market Streets, Galveston, TX 77555, USA
| | | |
Collapse
|
48
|
Gontero P, Banisadr S, Frea B, Brausi M. Metastasis markers in bladder cancer: a review of the literature and clinical considerations. Eur Urol 2005; 46:296-311. [PMID: 15306099 DOI: 10.1016/j.eururo.2004.04.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2004] [Indexed: 12/22/2022]
Abstract
Cancer invasion and metastasis develop through a sequence of processes involving loss of cell-cell and cell-matrix adhesions, proteolysis and induction of angiogenesis. We reviewed the current literature on the molecules that have been shown to play a significant role in these three steps of metastatisation in bladder cancer (BC) cells and their host microenvironment. Particular emphasis was given to markers that are assessable through immunohistochemistry and for which an additional prognostic value over the TNM variables has been recognized, in order to identify a subset of tumour markers readily available for application in daily clinical practice. We conclude that markers such as E-cadherin, Sialosyl-LeX, laminin, collagen IV, TSP-1 and MVD are useful prognostic markers, alpha, beta, and gamma catenin, MMP-2 and -9, uPAR, PD-ECGF and Bfgf can be considered potentially useful, while research on CD44, MMP-1 and -3, uPA, cathepsin D and VEGF has proved inconclusive. Further research in this field should concentrate on the molecules listed in the first group.
Collapse
Affiliation(s)
- Paolo Gontero
- Department of Urology, Università del Piemonte Orientale, Novara, Italy.
| | | | | | | |
Collapse
|
49
|
Calaluce R, Bearss DJ, Barrera J, Zhao Y, Han H, Beck SK, McDaniel K, Nagle RB. Laminin-5 beta3A expression in LNCaP human prostate carcinoma cells increases cell migration and tumorigenicity. Neoplasia 2004; 6:468-79. [PMID: 15548355 PMCID: PMC1531651 DOI: 10.1593/neo.03499] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2003] [Revised: 05/24/2004] [Accepted: 06/01/2004] [Indexed: 01/13/2023]
Abstract
Interactions between extracellular matrix proteins and prostate carcinoma cells change dramatically during prostate tumor progression. We have concentrated on two key modifications that occur in the hemidesmosome in prostate carcinoma: loss of laminin-5 protein expression and altered basal cell polarity of the alpha6beta4 integrin. We previously demonstrated two cell line-specific isoforms (beta3A and beta3B) of the LAMB3 message. Cells expressing only the beta3B isoform did not translate the beta3 protein and were unable to assemble the laminin-5 trimer. One such cell line, LNCaP, was selected to determine whether restoration of the laminin-5 beta3A isoform would cause expression of a functional laminin-5 beta3 chain, assembly and secretion of the laminin-5 trimer, and reversion to a non-neoplastic phenotype. Laminin-5 beta3A cDNA was cloned and stably transfected into LNCaP cells. We observed the restoration of the beta3 protein, but a laminin-5 trimer was not secreted. Moreover, increased cell migration was demonstrated, and tumorigenicity was increased in SCID mice. A microarray analysis, performed between transfected and nontransfected LNCaP cells, showed most changing genes to be associated with signal transduction. The beta3 chain of laminin-5 may thus play an important role in signal transduction, which may enhance cell motility and tumorigenesis.
Collapse
Affiliation(s)
- Robert Calaluce
- Arizona Cancer Center, University of Arizona Health Sciences Center, Tucson, AZ, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
During human prostate cancer progression, the majority of normally expressed integrins are suppressed with the exception of the alpha6, alpha3, and beta1 integrins. We have shown that in prostate cancer, the alpha6 integrin is found paired with the beta1 integrin and that a novel form of the alpha6 integrin that lacks a large portion of the extracellular domain (alpha6p) exists. The alpha6pbeta1 integrin is found in human prostate cancer tissue specimens as well as tissue culture cell lines and is formed on the cell surface. This review discusses the mechanism of alpha6pbeta1 production and the potential functions of this integrin variant. Our current working model predicts that the alpha6pbeta1 integrin maintains the intracellular cytoskeletal connections associated with the heterodimer while allowing for an alteration in cell adhesion. The mechanism provides a selective advantage for cancer cell metastasis.
Collapse
Affiliation(s)
| | - Anne E. Cress
- Correspondence to: Anne E. Cress, PhD, The Arizona Cancer Center, The University of Arizona, 1501 N. Campbell Ave., Tucson, AZ 85724. E-mail:
| |
Collapse
|