101
|
Integrin α3β1 regulates tumor cell responses to stromal cells and can function to suppress prostate cancer metastatic colonization. Clin Exp Metastasis 2012; 30:541-52. [PMID: 23224938 DOI: 10.1007/s10585-012-9558-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 11/24/2012] [Indexed: 10/27/2022]
Abstract
Integrin α3β1 promotes tumor cell adhesion, migration, and invasion on laminin isoforms, and several clinical studies have indicated a correlation between increased tumoral α3β1 integrin expression and tumor progression, metastasis, and poor patient outcomes. However, several other clinical and experimental studies have suggested that α3β1 can possess anti-metastatic activity in certain settings. To help define the range of α3β1 functions in tumor cells in vivo, we used RNAi to silence the α3 integrin subunit in an aggressive, in vivo-passaged subline of PC-3 prostate carcinoma cells. Loss of α3 integrin impaired adhesion and proliferation on the α3β1 integrin ligand, laminin-332 in vitro. Despite these deficits in vitro, the α3-silenced cells were significantly more aggressive in a lung colonization model in vivo, with a substantially increased rate of tumor growth that significantly reduced survival. In contrast, silencing the related α6 integrin subunit delayed metastatic growth in vivo. The increased colonization of α3-silenced tumor cells in vivo was recapitulated in 3D collagen co-cultures with lung fibroblasts or pre-osteoblast-like cells, where α3-silenced cells showed dramatically enhanced growth. The increased response of α3-silenced tumor cells to stromal cells in co-culture could be reproduced by fibroblast conditioned medium, which contains one or more heparin-binding factors that selectively favor the growth of α3-silenced cells. Our new data suggest a scenario in which α3β1 regulates tumor-host interactions within the metastatic tumor microenvironment to limit growth, providing some of the first direct evidence that specific loss of α3 function in tumor cells can have pro-metastatic consequences in vivo.
Collapse
|
102
|
Hudak L, Tezeeh P, Wedel S, Makarević J, Juengel E, Tsaur I, Bartsch G, Wiesner C, Haferkamp A, Blaheta RA. Low dosed interferon alpha augments the anti-tumor potential of histone deacetylase inhibition on prostate cancer cell growth and invasion. Prostate 2012; 72:1719-35. [PMID: 22473339 DOI: 10.1002/pros.22525] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 03/07/2012] [Indexed: 11/08/2022]
Abstract
We evaluated whether low-dosed interferon alpha (IFNa) may augment the anti-tumor potential of the histone deacetylase (HDAC)-inhibitor valproic acid (VPA) on prostate cancer cells in vitro and in vivo. PC-3, DU-145, or LNCaP prostate cancer cells were treated with VPA (1 mM), IFNa (200 U/ml), or with the VPA-IFNa combination. Tumor cell growth, cell cycle progression, and cell cycle regulating proteins were then investigated by the MTT assay, flow cytometry, and western blotting. Tumor cell adhesion to endothelium or to immobilized extracellular matrix proteins, as well as migratory properties of the cells, were evaluated. Integrin α and β adhesion molecules and alterations of cell signaling pathways were analyzed. Finally, effects of the drug treatment on prostate cancer growth in vivo were determined in the NOD/SCID mouse model. VPA reduced tumor cell adhesion, migration, and growth in vitro. A much stronger anti-cancer potential was evoked by the VPA-IFNa combination, although IFNa in itself did not block growth or adhesion. The same effect was seen when tumor growth was evaluated in vivo. Molecular analysis revealed distinct elevation of histone H3 acetylation caused by VPA which was further up-regulated by VPA-IFNa, whereas IFNa alone did not alter H3 acetylation. The combinatorial benefit became obvious in Akt phosphorylation, p21 and p27 and integrin α1, α3, and β1 expression. Application of low-dosed IFNa to a VPA based regimen profoundly boosts the anti-tumor properties of VPA. The combined use of VPA and low-dosed IFNa may therefore be an innovative option in treating advanced prostate cancer.
Collapse
Affiliation(s)
- Lukasz Hudak
- Department of Urology, Goethe-University, Frankfurt am Main, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
103
|
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: 14] [Impact Index Per Article: 1.2] [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
|
104
|
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: 4.2] [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
|
105
|
Liu J, Huang J, He Y, Liu J, Liao B, Liao G. Genetic variants in the integrin gene predicted microRNA-binding sites were associated with the risk of prostate cancer. Mol Carcinog 2012; 53:280-5. [PMID: 23065910 DOI: 10.1002/mc.21973] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Revised: 09/03/2012] [Accepted: 09/22/2012] [Indexed: 02/05/2023]
Abstract
microRNAs (miRNA) silence target genes through Watson-Crick based binding to the 3'untranslated regions (3'UTR). Thus, polymorphisms in the miRNA-binding sites may disrupt this process and play a potential role in cancer pathogenesis. Integrins have been implicated in the genesis and development of many tumors. This study was designed to evaluate the association between five SNP loci in predicted miRNA-binding sites in five integrin genes and prostate cancer occurrence and prognosis to provide data for screening high-risk Chinese Han individuals. These five polymorphisms were genotyped by using the high-resolution melting method (HRM) in 347 Chinese Han prostate cancer patients with long-time follow-up together with 367 age-matched healthy controls. GC carriers of rs11902171 in ITGAv were associated with a decreased risk of prostate cancer (OR 0.57, 95% CI 0.35-0.93). However, no significant difference was detected in genotype distributions of the five SNP loci in the progression-free survival time of prostate cancer. The ITGAv gene SNP rs11902171 may be potentially associated with the risk of prostate cancer.
Collapse
Affiliation(s)
- Jiaming Liu
- Department of Urology, West China School of Medicine/West China Hospital, Sichuan University, Sichuan Province, P.R., China
| | | | | | | | | | | |
Collapse
|
106
|
Trerotola M, Li J, Alberti S, Languino LR. Trop-2 inhibits prostate cancer cell adhesion to fibronectin through the β1 integrin-RACK1 axis. J Cell Physiol 2012; 227:3670-7. [PMID: 22378065 DOI: 10.1002/jcp.24074] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Trop-2 is a transmembrane glycoprotein upregulated in several human carcinomas, including prostate cancer (PrCa). Trop-2 has been suggested to regulate cell-cell adhesion, given its high homology with the other member of the Trop family, Trop-1/EpCAM, and its ability to bind the tight junction proteins claudin-1 and claudin-7. However, a role for Trop-2 in cell adhesion to the extracellular matrix has never been postulated. Here, we show for the first time that Trop-2 expression in PrCa cells correlates with their aggressiveness. Using either shRNA-mediated silencing of Trop-2 in cells that endogenously express it, or ectopic expression of Trop-2 in cells that do not express it, we show that Trop-2 inhibits PrCa cell adhesion to fibronectin (FN). In contrast, expression of another transmembrane receptor, α(v) β(5) integrin, does not affect cell adhesion to this ligand. We find that Trop-2 does not modulate either protein or activation levels of the prominent FN receptors, β(1) integrins, but acts through increasing β(1) association with the adaptor molecule RACK1 and redistribution of RACK1 to the cell membrane. As a result of Trop-2 expression, we also observe activation of Src and FAK, known to occur upon β(1) -RACK1 interaction. These enhanced Src and FAK activities are not mediated by changes in either the activity of IGF-IR, which is known to bind RACK1, or IGF-IR's ability to associate with β(1) integrins. In summary, our data demonstrate that the transmembrane receptor Trop-2 is a regulator of PrCa cell adhesion to FN through activation of the β(1) integrin-RACK1-FAK-Src signaling axis.
Collapse
Affiliation(s)
- Marco Trerotola
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | |
Collapse
|
107
|
Corn PG. The tumor microenvironment in prostate cancer: elucidating molecular pathways for therapy development. Cancer Manag Res 2012; 4:183-93. [PMID: 22904640 PMCID: PMC3421469 DOI: 10.2147/cmar.s32839] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mechanisms leading to the development of virulent prostate cancer are not confined to the cancer epithelial cell, but also involve the tumor microenvironment. Multiple signaling pathways exist between epithelial cells, stromal cells, and the extracellular matrix to support tumor progression from the primary site to regional lymph nodes and distant metastases. Prostate cancers preferentially metastasize to the skeleton, prompting considerable research effort into understanding the unique interaction between prostate cancer epithelial cells and the bone microenvironment. This effort has led to the discovery that signaling pathways involved in normal prostate and bone development become dysregulated in cancer. These pathways stimulate excessive cell growth and neovascularization, impart more invasive properties to epithelial cells, weaken antitumor immune surveillance, and promote the emergence of castrate-resistant disease. An improved understanding of the complex relationship between cancer epithelial cells and the organ-specific microenvironments with which they interact has created a powerful opportunity to develop novel therapies.
Collapse
Affiliation(s)
- Paul G Corn
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
108
|
β1 integrin deletion enhances progression of prostate cancer in the TRAMP mouse model. Sci Rep 2012; 2:526. [PMID: 22829980 PMCID: PMC3402831 DOI: 10.1038/srep00526] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 07/06/2012] [Indexed: 01/22/2023] Open
Abstract
β1 integrin regulates the response of both normal and cancer cells to their local environment. Although mis-localised in prostate cancer, the role β1 integrin plays in prostate development and carcinogenesis remains unknown. To assess the role of β1 integrin in vivo, we conditionally deleted β1 integrin from prostate epithelium and subsequently crossed these mice to the TRAMP prostate carcinogenesis model. Deletion of β1 integrin following castration and subsequent androgen supplementation resulted in an expansion of the p63-positive basal cell population and decreased differentiation. Consistent with these findings, deletion of β1 integrin in TRAMP mice decreased animal survival, decreased retention of normal prostate morphology, increased the percentage of tissue with poorly differentiated carcinoma, and increased cell proliferation. This study demonstrates that β1 integrin regulates several aspects of normal prostate development and in contrast to its role in several other tissues, its loss is associated with increased rates of prostate tumour progression.
Collapse
|
109
|
Colombel M, Eaton CL, Hamdy F, Ricci E, van der Pluijm G, Cecchini M, Mege-Lechevallier F, Clezardin P, Thalmann G. Increased expression of putative cancer stem cell markers in primary prostate cancer is associated with progression of bone metastases. Prostate 2012; 72:713-20. [PMID: 21882211 DOI: 10.1002/pros.21473] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 07/13/2011] [Indexed: 11/10/2022]
Abstract
BACKGROUND A number of putative stem cell markers have been associated with aggressiveness of prostate cancer, including alpha 2 and alpha 6 integrin and c-met. The study aimed to test the hypothesis that the development of bone metastasis correlates with the proportion of prostate cancer stem cell-like cells present in the primary tumor. METHODS Prostate tissue samples were obtained from patients with high-risk prostatic adenocarcinoma. Prostate cancer tumor tissue samples underwent immunohistochemical staining for alpha 2 and alpha 6 integrin and c-met; positive and negative controls were included. Samples were scored as positive if >5% of cells within the sample stained positively. Survival and bone metastasis-free survival curves on the patient cohort were estimated by the actuarial method of Kaplan-Meier. RESULTS A total of 62 patients were included in the study. Bone metastases progression rate was 46% at 105 months with a median time of 46 months (95% CI: 1-62.5 months); prostate cancer-specific survival was 33% at 122 months with a median survival time of 69.4 months (95% CI: 63.5-109.4 months). Survival curves show that c-met-, alpha 2, and alpha 6 integrin-positive tumors were positively associated with the occurrence of bone metastasis-free survival. There was a higher level of significance when at least c-met and either alpha 2 or alpha 6 integrin was positive. CONCLUSION It can be concluded that percentage of stem cell-like prostate cancer cells has a prognostic impact especially on the risk of metastatic bone progression.
Collapse
Affiliation(s)
- Marc Colombel
- Service d'Urologie et Chirurgie de la Transplantation, Université Lyon 1, Lyon, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Saxena P, Trerotola M, Wang T, Li J, Sayeed A, VanOudenhove J, Adams DS, FitzGerald TJ, Altieri DC, Languino LR. PSA regulates androgen receptor expression in prostate cancer cells. Prostate 2012; 72:769-76. [PMID: 21956655 PMCID: PMC3404455 DOI: 10.1002/pros.21482] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 08/22/2011] [Indexed: 12/31/2022]
Abstract
BACKGROUND Prostate-specific antigen (PSA) is a pivotal downstream target gene of the androgen receptor (AR), and a serum biomarker to monitor prostate cancer (PrCa) progression. It has been reported that PSA transactivates AR, but the mechanistic requirements of this response have not been investigated. METHODS We studied the localization of PSA, AR, and Src in intracellular compartments of synthetic androgen (R1881)-stimulated LNCaP and C4-2B PrCa cells, using immunofluorescence and subcellular fractionation approaches. We also investigated the effect of downregulation of PSA on AR expression by immunoblotting and real-time PCR using short hairpin RNA (shRNA) and small interfering RNA (siRNA). Src activity was analyzed by immunoblotting. RESULTS R1881 stimulation induced nuclear localization of both PSA and AR in LNCaP and C4-2B PrCa cells as well as increased phosphorylation of Src. Stable shRNA or transient siRNA knockdown of PSA resulted in reduced AR protein levels as well as AR mRNA levels in C4-2B cells. Similar to C4-2B cells, ablation of AR levels upon silencing of PSA was also confirmed in VCaP cells, another androgen-independent cell line. Silencing of PSA did not cause significant changes in Src activation; besides, Src regulation by integrins did not appear to affect AR transcriptional activity. CONCLUSIONS PSA localizes to nuclei of androgen-stimulated PrCa cells, and controls AR mRNA and protein levels. This regulatory loop is specific for PSA, does not involve known AR activators such as Src and AKT, and may contribute to AR signaling under conditions of increasing PSA levels in patients.
Collapse
Affiliation(s)
- Parmita Saxena
- Department of Cancer Biology, Prostate Cancer Discovery and Development Program, University of Massachusetts Medical School, Worcester, MA 01605
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester MA 01609
| | - Marco Trerotola
- Department of Cancer Biology, Prostate Cancer Discovery and Development Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Tao Wang
- Department of Cancer Biology, Prostate Cancer Discovery and Development Program, University of Massachusetts Medical School, Worcester, MA 01605
- Department of Radiation Oncology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Jing Li
- Department of Cancer Biology, Prostate Cancer Discovery and Development Program, University of Massachusetts Medical School, Worcester, MA 01605
| | - Aejaz Sayeed
- Department of Cancer Biology, Prostate Cancer Discovery and Development Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Jennifer VanOudenhove
- Department of Cancer Biology, Prostate Cancer Discovery and Development Program, University of Massachusetts Medical School, Worcester, MA 01605
| | - Dave S. Adams
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester MA 01609
| | - Thomas J. FitzGerald
- Department of Radiation Oncology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Dario C. Altieri
- Prostate Cancer Discovery and Development Program, The Wistar Institute Cancer Center, Philadelphia, PA19104
| | - Lucia R. Languino
- Department of Cancer Biology, Prostate Cancer Discovery and Development Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
- Correspondence to: Lucia R. Languino Ph.D., Department of Cancer Biology, Thomas Jefferson University, 233 South 10 Street, Philadelphia, PA 19107. Phone: 215.503.3442. Fax: 215.503.1607.
| |
Collapse
|
111
|
Riggs KA, Hasan N, Humphrey D, Raleigh C, Nevitt C, Corbin D, Hu C. Regulation of integrin endocytic recycling and chemotactic cell migration by syntaxin 6 and VAMP3 interaction. J Cell Sci 2012; 125:3827-39. [PMID: 22573826 DOI: 10.1242/jcs.102566] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Integrins are the primary receptors of cells adhering to the extracellular matrix, and play key roles in various cellular processes including migration, proliferation and survival. The expression and distribution of integrins at the cell surface is controlled by endocytosis and recycling. The present study examines the function of syntaxin 6 (STX6), a t-SNARE located in the trans-Golgi network, in integrin trafficking. STX6 is overexpressed in many types of human cancer. We show that depletion of STX6 inhibits chemotactic cell migration and the delivery of the laminin receptor α3β1 integrin to the cell surface, whereas STX6 overexpression stimulates chemotactic cell migration, integrin delivery, and integrin-initiated activation of focal adhesion kinase. These data indicate that STX6 plays a rate-limiting role in cell migration and integrin trafficking. In STX6-depleted cells, α3β1 integrin is accumulated in recycling endosomes that contain the v-SNARE VAMP3. Importantly, we show that STX6 and VAMP3 form a v-/t-SNARE complex, VAMP3 is required in α3β1 integrin delivery to the cell surface, and endocytosed α3β1 integrin traffics to both VAMP3 and STX6 compartments. Collectively, our data suggest a new integrin trafficking pathway in which endocytosed integrins are transported from VAMP3-containing recycling endosomes to STX6-containing trans-Golgi network before being recycled to the plasma membrane.
Collapse
Affiliation(s)
- Krista A Riggs
- Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | | | | | | | | | | | | |
Collapse
|
112
|
Garlick DS, Li J, Sansoucy B, Wang T, Griffith L, FitzGerald TJ, Butterfield J, Charbonneau B, Violette SM, Weinreb PH, Ratliff TL, Liao CP, Roy-Burman P, Vietri M, Lian JB, Stein GS, Altieri DC, Languino LR. α(V)β(6) integrin expression is induced in the POET and Pten(pc-/-) mouse models of prostatic inflammation and prostatic adenocarcinoma. Am J Transl Res 2012; 4:165-174. [PMID: 22611469 PMCID: PMC3353537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 04/06/2012] [Indexed: 06/01/2023]
Abstract
Chronic inflammation is proposed to prime the development of prostate cancer. However, the mechanisms of prostate cancer initiation and development are not completely understood. The α(v)β(6) integrin has been shown to play a role in epithelial development, wound healing and some epithelial cancers [1, 2]. Here, we investigate the expression of α(v)β(6) in mouse models of prostatic inflammation and prostate cancer to establish a possible relationship between inflammation of the prostate, α(v)β(6) expression and the progression of prostate cancer. Using immunohistochemical techniques, we show expression of α(v)β(6) in two in vivo mouse models; the Pten(pc)-/- model containing a prostate- specific Pten tumor suppressor deletion that causes cancer, and the prostate ovalbumin-expressing transgenic (POET) inflammation mouse model. We show that the α(v)β(6) integrin is induced in prostate cancer and inflammation in vivo in these two mouse models. α(v)β(6) is expressed in all the mice with cancer in the Pten(pc-/-) model but not in age-matched wild-type mice. In the POET inflammation model, α(v)β(6) is expressed in mice injected with activated T-cells, but in none of the control mice. In the POET model, we also used real time PCR to assess the expression of Transforming Growth Factor Beta 1 (TGFβ1), a factor in inflammation that is activated by α(v)β(6). In conclusion, through in vivo evidence, we conclude that α(v)β(6) integrin may be a crucial link between prostatic inflammation and prostatic adenocarcinoma.
Collapse
Affiliation(s)
- David S Garlick
- Department of Cancer Biology and Cancer Center, University of Massachusetts Medical SchoolWorcester, MA
| | - Jing Li
- Department of Cancer Biology and Cancer Center, University of Massachusetts Medical SchoolWorcester, MA
| | - Brian Sansoucy
- Department of Cancer Biology and Cancer Center, University of Massachusetts Medical SchoolWorcester, MA
| | - Tao Wang
- Department of Radiation Oncology, University of Massachusetts Medical SchoolWorcester, MA
| | - Leeanne Griffith
- Prostate Cancer Discovery and Development Program, Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson UniversityPhiladelphia, PA.
| | - TJ FitzGerald
- Department of Radiation Oncology, University of Massachusetts Medical SchoolWorcester, MA
| | - Julie Butterfield
- Department of Cancer Biology and Cancer Center, University of Massachusetts Medical SchoolWorcester, MA
| | - Bridget Charbonneau
- Purdue University Center for Cancer Research, Department of Comparative Pathobiology, School of Veterinary Medicine, Purdue UniversityWest Lafayette, IN
| | | | | | - Timothy L Ratliff
- Purdue University Center for Cancer Research, Department of Comparative Pathobiology, School of Veterinary Medicine, Purdue UniversityWest Lafayette, IN
| | - Chun-Peng Liao
- Department of Pathology, Keck School of Medicine, University of Southern CaliforniaLos Angeles, CA
| | - Pradip Roy-Burman
- Department of Pathology, Keck School of Medicine, University of Southern CaliforniaLos Angeles, CA
| | - Michele Vietri
- Department of Cancer Biology and Cancer Center, University of Massachusetts Medical SchoolWorcester, MA
| | - Jane B Lian
- Prostate Cancer Discovery and Development Program, Department of Cell Biology and Cancer Center, University of Massachusetts Medical SchoolWorcester, MA
| | - Gary S Stein
- Prostate Cancer Discovery and Development Program, Department of Cell Biology and Cancer Center, University of Massachusetts Medical SchoolWorcester, MA
| | - Dario C Altieri
- Prostate Cancer Discovery and Development Program, The Wistar Institute Cancer CenterPhiladelphia, PA
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson UniversityPhiladelphia, PA.
| |
Collapse
|
113
|
Jia D, Entersz I, Butler C, Foty RA. Fibronectin matrix-mediated cohesion suppresses invasion of prostate cancer cells. BMC Cancer 2012; 12:94. [PMID: 22433434 PMCID: PMC3359283 DOI: 10.1186/1471-2407-12-94] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 03/20/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Invasion is an important early step in the metastatic cascade and is the primary cause of death of prostate cancer patients. In order to invade, cells must detach from the primary tumor. Cell-cell and cell-ECM interactions are important regulators of cohesion--a property previously demonstrated to mediate cell detachment and invasion. The studies reported here propose a novel role for α5β1 integrin--the principle mediator of fibronectin matrix assembly (FNMA)--as an invasion suppressor of prostate cancer cells. METHODS Using a combination of biophysical and cell biological methods, and well-characterized prostate cancer cell lines of varying invasiveness, we explore the relationship between cohesion, invasiveness, and FNMA. RESULTS We show that cohesion is inversely proportional to invasive capacity. We also show that more invasive cells express lower levels of α5β1 integrin and lack the capacity for FNMA. Cells were generated to over-express either wild-type α5 integrin or an integrin in which the cytoplasmic domain of α5 was replaced with that of α2. The α2 construct does not promote FNMA. We show that only wild-type α5 integrin promotes aggregate compaction, increases cohesion, and reduces invasion of the more aggressive cells, and that these effects can be blocked by the 70-kDa fibronectin fragment. CONCLUSIONS We propose that restoring capacity for FNMA in deficient cells can increase tumor intercellular cohesion to a point that significantly reduces cell detachment and subsequent invasion. In prostate cancer, this could be of therapeutic benefit by blocking an early key step in the metastatic cascade.
Collapse
Affiliation(s)
- Dongxuan Jia
- Department of Surgery, UMDNJ-Robert Wood Johnson Medical School, 125 Paterson Street, CAB 7319, New Brunswick, NJ 08901, USA
| | | | | | | |
Collapse
|
114
|
Abstract
Tumor progenitor cells represent a population of drug-resistant cells that can survive conventional chemotherapy and lead to tumor relapse. However, little is known of the role of tumor progenitors in prostate cancer metastasis. The studies reported herein show that the CXCR4/CXCL12 axis, a key regulator of tumor dissemination, plays a role in the maintenance of prostate cancer stem-like cells. The CXCL4/CXCR12 pathway is activated in the CD44+/CD133+ prostate progenitor population and affects differentiation potential, cell adhesion, clonal growth and tumorigenicity. Furthermore, prostate tumor xenograft studies in mice showed that a combination of the CXCR4 receptor antagonist AMD3100, which targets prostate cancer stem-like cells, and the conventional chemotherapeutic drug Taxotere, which targets the bulk tumor, is significantly more effective in eradicating tumors as compared to monotherapy.
Collapse
|
115
|
Sayeed A, Alam N, Trerotola M, Languino LR. Insulin-like growth factor 1 stimulation of androgen receptor activity requires β(1A) integrins. J Cell Physiol 2012; 227:751-8. [PMID: 21465482 DOI: 10.1002/jcp.22784] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Despite the findings that β1 integrins play a vital role in the regulation of cell proliferation and survival, the mechanisms through which they operate and lead to cancer progression remain elusive. Previously, our laboratory has shown that β(1A) integrins support insulin-like growth factor 1 (IGFI)-mediated mitogenic and transforming activities. Here, we report that β(1A) integrins regulate basal levels of IGF-IR, although they are not critical for maintaining cancer cell morphology. Upon transfection of β(1A) siRNA and consequent downregulation of IGF-IR, we show inhibition of anchorage-independent growth of prostate cancer cells, a function which is dependent on IGF-IR expression. In addition, we demonstrate that IGFI-mediated activation of androgen receptor (AR), known to occur in prostate cancer cells, requires expression of β(1A) integrins as evaluated by luciferase reporter assays and immunoblotting analysis. Since β(1A) integrin levels are increased by R1881 or dihydrotestosterone (DHT), our results imply that β(1A) integrins support an androgen-enhanced feedback loop that regulates the expression of IGF-IR. β(1A) integrins also regulate inducible levels of IGF-IR in cells stimulated by androgen or by a combination of androgen and IGFI, as evaluated by flow cytometric analysis and immunoblotting. Furthermore, upon transfection of β(1A) siRNA and consequent downregulation of IGF-IR, neither activation of AKT, an effector of IGF-IR, nor AR levels are affected. We conclude that β(1A) integrin expression is critical for maintaining the regulatory crosstalk between IGF-IR and AR.
Collapse
Affiliation(s)
- Aejaz Sayeed
- Department of Cancer Biology, Prostate Cancer Discovery and Development Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
116
|
Liao CP, Adisetiyo H, Liang M, Roy-Burman P. Cancer stem cells and microenvironment in prostate cancer progression. Discov Oncol 2011; 1:297-305. [PMID: 21761361 DOI: 10.1007/s12672-010-0051-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
For a study of interactions between the cancer-associated fibroblasts (CAFs) and the putative prostate cancer stem cells (CSCs), we used a conditional Pten deletion mouse model of prostatic adenocarcinoma to isolate both CAF cultures and CSC-enriched cell fractions from the primary tumors. The CSC subpopulation exhibited a collective phenotype of Lin(-)/SCA-1(hi)/CD49f(hi)/p63(hi)/CK5(hi)/AR(lo)/CK18(lo)/Survivin(hi)/Runx2(hi) and contained cells with the ability to both self-renew and differentiate into basal and luminal cells in vitro. The spheroids generated from the CSC-enriched subpopulation mimicked the glandular structures that could be produced from a similarly isolated cell fraction from the normal mouse prostate. The efficiency of spheroid formation was found to be influenced differentially by the nature of the fibroblasts that were co-cultured in the 3-D system. The growth and differentiation properties of the CSCs were significantly more enhanced by factors released from CAFs relative to normal prostate fibroblasts (NPFs). Additionally, increased commitment to differentiation to the luminal cell lineage was noted when CAFs were present. When CSCs admixed with either CAFs or NPFs were examined for formation of prostatic glandular structures in renal grafts in vivo, the lesions formed were generally more in numbers in the presence of CAFs than NPFs. Furthermore, lesions formed with CAFs often displayed tumor-like complex histopathology and contained increased numbers of proliferating cells. Taken together, the results suggested that the CAFs in the prostate tumor microenvironment can contribute to the biologic properties of the CSCs and by this account may play a major role in prostate tumorigenesis and progression. Thus, it would be important now to identify the paracrine and/or juxtacrine factors that are responsible for the stimulation of the cancer stem cells.
Collapse
Affiliation(s)
- Chun-Peng Liao
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, 2011 Zonal Avenue, Los Angeles, CA 90033, USA
| | | | | | | |
Collapse
|
117
|
Wang Q, Bailey CG, Ng C, Tiffen J, Thoeng A, Minhas V, Lehman ML, Hendy SC, Buchanan G, Nelson CC, Rasko JEJ, Holst J. Androgen receptor and nutrient signaling pathways coordinate the demand for increased amino acid transport during prostate cancer progression. Cancer Res 2011; 71:7525-36. [PMID: 22007000 DOI: 10.1158/0008-5472.can-11-1821] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
L-Type amino acid transporters such as LAT1 and LAT3 mediate the uptake of essential amino acids. Here, we report that prostate cancer cells coordinate the expression of LAT1 and LAT3 to maintain sufficient levels of leucine needed for mTORC1 signaling and cell growth. Inhibiting LAT function was sufficient to decrease cell growth and mTORC1 signaling in prostate cancer cells. These cells maintained levels of amino acid influx through androgen receptor-mediated regulation of LAT3 expression and ATF4 regulation of LAT1 expression after amino acid deprivation. These responses remained intact in primary prostate cancer, as indicated by high levels of LAT3 in primary disease, and by increased levels of LAT1 after hormone ablation and in metastatic lesions. Taken together, our results show how prostate cancer cells respond to demands for increased essential amino acids by coordinately activating amino acid transporter pathways vital for tumor outgrowth.
Collapse
Affiliation(s)
- Qian Wang
- Origins of Cancer Laboratory, Centenary Institute, Newtown, NSW, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Wedel S, Hudak L, Seibel JM, Juengel E, Oppermann E, Haferkamp A, Blaheta RA. Critical analysis of simultaneous blockage of histone deacetylase and multiple receptor tyrosine kinase in the treatment of prostate cancer. Prostate 2011; 71:722-35. [PMID: 20954195 DOI: 10.1002/pros.21288] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Accepted: 09/07/2010] [Indexed: 11/09/2022]
Abstract
BACKGROUND The concept of molecular tumor targeting might be an innovative option to treat advanced prostate cancer. We analyzed the effect of combining the multiple receptor tyrosine kinase inhibitor AEE788 and the histone deacetylase (HDAC) inhibitor valproic acid (VPA) on adhesion and growth properties of prostate cancer cell lines. METHODS PC-3, DU-145, and LNCaP cells were treated with AEE788, VPA or with an AEE788-VPA combination, and cell cycle progression investigated. Furthermore, tumor cell adhesion to vascular endothelium or to immobilized extracellular matrix proteins was evaluated, and integrin α and β subtypes were analyzed. Finally, effects of drug treatment on cell signaling pathways were determined. RESULTS AEE788 moderately and VPA strongly reduced tumor cell adhesion and growth. VPA impaired cell cycle progression and altered the expression level of the cell cycle regulating proteins cdk1, cdk2, cdk4, cyclin B, D1, cyclin E, p21, and p27. VPA also acted on the membranous, cytoplasmic, and gene expression pattern of various integrin α and β subtypes. AEE788 acted likewise, but more moderately. Combining AEE788 and VPA did not result in an additive anti-tumor effect. Signaling analysis revealed that the EGFr downstream target Akt was similarly modified in the presence of VPA or the VPA-AEE788 combination, but not influenced by AEE788 alone. CONCLUSIONS The AEE788-VPA combination has no advantage over VPA monotreatment in vitro. The non-responsiveness of Akt
Collapse
Affiliation(s)
- Steffen Wedel
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | | | | | | | | | | | | |
Collapse
|
119
|
Neal CL, Mckeithen D, Odero-Marah VA. Snail negatively regulates cell adhesion to extracellular matrix and integrin expression via the MAPK pathway in prostate cancer cells. Cell Adh Migr 2011; 5:249-57. [PMID: 21478672 DOI: 10.4161/cam.5.3.15618] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Snail transcription factor induces epithelial-mesenchymal transition (EMT) in which the epithelial cells downregulate cell-cell adhesion genes such as E-Cadherin and upregulate mesenchymal genes such as vimentin, leading to increased invasion and migration. Very little is known about the role of Snail in cellular adhesion to the extracellular matrix. We hypothesized that Snail will lead to decreased cellular adhesion to fibronectin and collagen I matrix through integrin regulation, concomitant with increased cell migration. Androgen-independent C4-2 cells, an aggressive subline of androgen-dependent LNCaP cells, exhibited decreased cell adhesion and increased cell migration on fibronectin and collagen I as compared to LNCaP cells, which was reversed by Snail knock down in C4-2 cells. ARCaP and LNCaP prostate cancer cells stably transfected with Snail displayed decreased adhesion and increased cell migration on fibronectin and collagen I as compared to control Neo-transfected cells, which was reversed by Snail knockdown. Flow cytometry analysis revealed a decrease in a5, a2 and b1 integrin expression in ARCaP Snail-transfected cells that was reversed in Snail knock down cells. We also observed an increase in ERK phosphorylation in ARCaP Snail-transfected cells as compared to control ARCaP-Neo cells, and inhibition of the MAPK pathway with UO126 inhibitor in ARCaP Snail-transfected cells abrogated Snail-mediated decrease in cell adhesion and reinduced a5, a2 and b1 integrin expression. Collectively, these studies define a new role for Snail transcription factor in cell adhesion to the ECM, which may be mediated by MAPK signaling, and may be crucial for cell detachment and subsequent metastasis.
Collapse
Affiliation(s)
- Corey L Neal
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
| | | | | |
Collapse
|
120
|
Components of cell-matrix linkage as potential new markers for prostate cancer. Cancers (Basel) 2011; 3:883-96. [PMID: 24212644 PMCID: PMC3756394 DOI: 10.3390/cancers3010883] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 02/12/2011] [Accepted: 02/17/2011] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer is one of the most common tumor diseases worldwide. Often being non-aggressive, prostate tumors in these cases do not need immediate treatment. However, about 20% of diagnosed prostate cancers tend to metastasize and require treatment. Existing diagnostic methods may fail to accurately recognize the transition of a dormant, non-aggressive tumor into highly malignant prostate cancer. Therefore, new diagnostic tools are needed to improve diagnosis and therapy of prostate carcinoma. This review evaluates existing methods to diagnose prostate carcinoma, such as the biochemical marker prostate-specific antigen (PSA), but also discusses the possibility to use the altered expression of integrins and laminin-332 in prostate carcinomas as diagnostic tools and therapeutic targets of prostate cancer.
Collapse
|
121
|
Karlou M, Tzelepi V, Efstathiou E. Therapeutic targeting of the prostate cancer microenvironment. Nat Rev Urol 2011; 7:494-509. [PMID: 20818327 DOI: 10.1038/nrurol.2010.134] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Solid tumors can be thought of as multicellular 'organs' that consist of a variety of cells as well as a scaffold of noncellular matrix. Stromal-epithelial crosstalk is integral to prostate cancer progression and metastasis, and androgen signaling is an important component of this crosstalk at both the primary and metastatic sites. Intratumoral production of androgen is an important mechanism of castration resistance and has been the focus of novel therapeutic approaches with promising results. Various other pathways are important for stromal-epithelial crosstalk and represent attractive candidate therapeutic targets. Hedgehog signaling has been associated with tumor progression, growth and survival, while Src family kinases have been implicated in tumor progression and in regulation of cancer cell migration. Fibroblast growth factors and transforming growth factor beta signaling regulate cell proliferation, apoptosis and angiogenesis in the prostate cancer microenvironment. Integrins mediate communication between the cell and the extracellular matrix, enhancing growth, migration, invasion and metastasis of cancer cells. The contribution of stromal-epithelial crosstalk to prostate cancer initiation and progression provides the impetus for combinatorial microenvironment-targeting strategies.
Collapse
Affiliation(s)
- Maria Karlou
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77230-1439, USA
| | | | | |
Collapse
|
122
|
Koivunen JT, Nissinen L, Juhakoski A, Pihlavisto M, Marjamäki A, Huuskonen J, Pentikäinen OT. Blockage of collagen binding to integrin α2β1: structure–activity relationship of protein–protein interaction inhibitors. MEDCHEMCOMM 2011. [DOI: 10.1039/c1md00089f] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
123
|
Desiniotis A, Kyprianou N. Significance of talin in cancer progression and metastasis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 289:117-47. [PMID: 21749900 DOI: 10.1016/b978-0-12-386039-2.00004-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Upon detachment from the extracellular matrix, tumor epithelial cells and tumor-associated endothelial cells are capable of overcoming anoikis, gain survival benefits, and hence contribute to the process of metastasis. The focal-adhesion complex formation recruits the association of key adaptor proteins such as FAK (focal-adhesion kinase). Vimentin, paxillin, and talin are responsible for mediating the interaction between the actin cytoskeleton and integrins. Talin is an early-recruited focal-adhesion player that is of structural and functional significance in mediating interactions with integrin cytoplasmic tails leading to destabilization of the transmembrane complex and resulting in rearrangements in the extracellular integrin compartments that mediate integrin activation. Talin-mediated integrin activation plays a definitive role in integrin-mediated signaling and induction of downstream survival pathways leading to protection from anoikis and consequently resulting in cancer progression to metastasis. We recently reported that talin expression is significantly increased in prostate cancer compared with benign and normal prostate tissue and that this overexpression correlates with progression to metastatic disease implicating a prognostic value for talin during tumor progression. At the molecular level, talin is functionally associated with enhanced survival and proliferation pathways and confers anoikis resistance and metastatic spread of primary tumor cells via activation of the Akt survival pathway. In this review, we discuss the growing evidence surrounding the value of talin as a prognostic marker of cancer progression to metastasis and as therapeutic target in advanced prostate cancer, as well as the current understanding of mechanisms regulating its signaling activity in cancer.
Collapse
Affiliation(s)
- Andreas Desiniotis
- Department of Surgery/Urology, and Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, USA
| | | |
Collapse
|
124
|
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: 2.0] [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
|
125
|
Caicedo-Carvajal CE, Shinbrot T, Foty RA. Alpha5beta1 integrin-fibronectin interactions specify liquid to solid phase transition of 3D cellular aggregates. PLoS One 2010; 5:e11830. [PMID: 20686611 PMCID: PMC2912296 DOI: 10.1371/journal.pone.0011830] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Accepted: 06/30/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Tissue organization during embryonic development and wound healing depends on the ability of cells on the one hand to exchange adhesive bonds during active rearrangement and on the other to become fixed in place as tissue homeostasis is reached. Cells achieve these contradictory tasks by regulating either cell-cell adhesive bonds, mediated by cadherins, or cell-extracellular matrix (ECM) connections, regulated by integrins. Integrin alpha5beta1 and soluble fibronectin (sFN) are key players in cell-ECM force generation and in ECM polymerization. Here, we explore the interplay between integrin alpha5beta1 and sFN and its influence on tissue mechanical properties and cell sorting behavior. METHODOLOGY/PRINCIPAL FINDINGS We generated a series of cell lines varying in alpha5beta1 receptor density. We then systematically explored the effects of different sFN concentrations on aggregate biomechanical properties using tissue surface tensiometry. We found previously unreported complex behaviors including the observation that interactions between fibronectin and integrin alpha5beta1 generates biphasic tissue cohesion profiles. Specifically, we show that at constant sFn concentration, aggregate cohesion increases linearly as alpha5beta1 receptor density is increased from low to moderate levels, producing a transition from viscoelastic-liquid to pseudo viscoelastic-solid behavior. However, further increase in receptor density causes an abrupt drop in tissue cohesion and a transition back to viscoelastic-liquid properties. We propose that this may be due to depletion of sFn below a critical value in the aggregate microenvironment at high alpha5beta1 levels. We also show that differential expression of alpha5beta1 integrin can promote phase-separation between cells. CONCLUSIONS/SIGNIFICANCE The interplay between alpha5-integrin and sFn contributes significantly to tissue cohesion and, depending on their level of expression, can mediate a shift from liquid to elastic behavior. This interplay represents a tunable level of control between integrins and the ECM that can influence tissue cohesion and other mechanical properties, which may translate to the specification of tissue structure and function. These studies provide insights into important biological processes such as embryonic development, wound healing, and for tissue engineering applications.
Collapse
Affiliation(s)
- Carlos E. Caicedo-Carvajal
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, United States of America
| | - Troy Shinbrot
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, United States of America
| | - Ramsey A. Foty
- Department of Surgery, University of Medicine and Dentistry, New Jersey-Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
126
|
Sroka IC, Anderson TA, McDaniel KM, Nagle RB, Gretzer MB, Cress AE. The laminin binding integrin alpha6beta1 in prostate cancer perineural invasion. J Cell Physiol 2010; 224:283-8. [PMID: 20432448 DOI: 10.1002/jcp.22149] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metastasizing prostate tumor cells invade along nerves innervating the encapsulated human prostate gland in a process known as perineural invasion. The extracellular matrix laminin class of proteins line the neural route and tumor cells escaping from the gland express the laminin binding integrin alpha6beta1 as a prominent cell surface receptor. Integrin alpha6beta1 promotes aggressive disease and supports prostate tumor cell metastasis to bone. Laminins and their integrin receptors are necessary for the development and maintenance of the peripheral nervous system, indicating the potential role for integrin receptors in directing prostate tumor cell invasion on nerves during perineural invasion.
Collapse
Affiliation(s)
- Isis C Sroka
- Department of Pharmacology, The University of Arizona, Tucson, Arizona, USA
| | | | | | | | | | | |
Collapse
|
127
|
Fredolini C, Meani F, Luchini A, Zhou W, Russo P, Ross M, Patanarut A, Tamburro D, Gambara G, Ornstein D, Odicino F, Ragnoli M, Ravaggi A, Novelli F, Collura D, D'Urso L, Muto G, Belluco C, Pecorelli S, Liotta L, Petricoin EF. Investigation of the ovarian and prostate cancer peptidome for candidate early detection markers using a novel nanoparticle biomarker capture technology. AAPS JOURNAL 2010; 12:504-18. [PMID: 20549403 DOI: 10.1208/s12248-010-9211-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Accepted: 06/01/2010] [Indexed: 11/30/2022]
Abstract
Current efforts to identify protein biomarkers of disease use mainly mass spectrometry (MS) to analyze tissue and blood specimens. The low-molecular-weight "peptidome" is an attractive information archive because of the facile nature by which the low-molecular-weight information freely crosses the endothelial cell barrier of the vasculature, which provides opportunity to measure disease microenvironment-associated protein analytes secreted or shed into the extracellular interstitium and from there into the circulation. However, identifying useful protein biomarkers (peptidomic or not) which could be useful to detect early detection/monitoring of disease, toxicity, doping, or drug abuse has been severely hampered because even the most sophisticated, high-resolution MS technologies have lower sensitivities than those of the immunoassays technologies now routinely used in clinical practice. Identification of novel low abundance biomarkers that are indicative of early-stage events that likely exist in the sub-nanogram per milliliter concentration range of known markers, such as prostate-specific antigen, cannot be readily detected by current MS technologies. We have developed a new nanoparticle technology that can, in one step, capture, concentrate, and separate the peptidome from high-abundance blood proteins. Herein, we describe an initial pilot study whereby the peptidome content of ovarian and prostate cancer patients is investigated with this method. Differentially abundant candidate peptidome biomarkers that appear to be specific for early-stage ovarian and prostate cancer have been identified and reveal the potential utility for this new methodology.
Collapse
|
128
|
Eaton CL, Colombel M, van der Pluijm G, Cecchini M, Wetterwald A, Lippitt J, Rehman I, Hamdy F, Thalman G. Evaluation of the frequency of putative prostate cancer stem cells in primary and metastatic prostate cancer. Prostate 2010; 70:875-82. [PMID: 20127735 DOI: 10.1002/pros.21121] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Tumour cells with a stem cell-like phenotype have recently been identified in prostate tumors and it has been suggested that this population may be responsible for the diversity of cell types within tumors and also for the initiation of metastases. These cells carry a number of defined markers: they are cd133 and cd44+ve and express high levels of alpha2beta1 integrin. In this study we have, for the first time, assessed matched primary and bone marrow biopsies from prostate cancer patients for the distribution of cells carrying these and a number of other putative stem cell markers. METHODS Eleven matched (primary and bone metastasis) specimens from prostate cancer patients were assessed for the presence of cd133, cd44, alpha2beta1 integrin, CXCR4, c-met, alpha6 integrin, and nestin using immunohistochemistry and stain intensity and distribution scored. RESULTS In the bone metastases, tumor cells staining positively for cd133 were detected at low frequency in approximately 50% of samples. Staining for nestin was confined to endothelium. Positive staining of tumor cells for the other antigens was present at variable frequency in >70% of metastases with the exception of CXCR4 which was absent from all but 2 specimens. Where positive staining of tumor cells was present in the metastasis, cells staining for each antigen were present in the matched primary with the exception of cd44 which was absent in all but 2/11 matched primary tissues. CONCLUSIONS In established metastases no single or combination of marker expression profiles identify the established metastatic phenotype, although cd44 expression was shown to be more frequent in metastases that in primary cancers.
Collapse
Affiliation(s)
- Colby L Eaton
- School of Medicine, University of Sheffield, Sheffield, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Goel HL, Underwood JM, Nickerson JA, Hsieh CC, Languino LR. Beta1 integrins mediate cell proliferation in three-dimensional cultures by regulating expression of the sonic hedgehog effector protein, GLI1. J Cell Physiol 2010; 224:210-7. [PMID: 20333644 DOI: 10.1002/jcp.22116] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The beta1 integrins play an important role in the modulation of cancer cell proliferation and tumor growth. We have previously shown that beta1 integrins associate with insulin-like growth factor type 1 receptor (IGF-IR) and regulate IGF-stimulated prostate cancer cell proliferation. In the present study, we find that downregulation of beta1 in prostate cancer cells inhibits IGF-IR and AKT activation. We also show that beta1 downregulation in two- and three-dimensional (3D) prostate cancer cell cultures significantly reduces expression of GLI1, a transcription factor known to be regulated by the IGF/AKT signaling pathway and to be a downstream effector of sonic hedgehog. Re-expression of GLI1 rescues the inhibitory effect of beta1 downregulation on prostate cancer cell proliferation in 3D cultures. We find that downregulation of beta1 significantly reduces surface expression of the associated alpha 5 integrin subunit. Our results indicate that the beta1/IGF-IR complex regulates expression of GLI1, which in turn promotes cancer cell proliferation in 3D cultures.
Collapse
Affiliation(s)
- Hira Lal Goel
- Department of Cancer Biology, Prostate Cancer Discovery and Development Program, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | | | | | |
Collapse
|
130
|
Nagrial A, Horvath L. Bone disease in prostate cancer. Asia Pac J Clin Oncol 2010; 6:3-4. [PMID: 20398032 DOI: 10.1111/j.1743-7563.2010.01281.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
131
|
Abstract
It is becoming increasingly clear that angiogenesis plays a crucial role in prostate cancer (CaP) survival, progression, and metastasis. Tumor angiogenesis is a hallmark of advanced cancers and an attractive treatment target in multiple solid tumors. By understanding the molecular basis of resistance to androgen withdrawal and chemotherapy in CaP, the rational design of targeted therapeutics is possible. This review summarizes the recent advancements that have improved our understanding of the role of angiogenesis in CaP metastasis and the potential therapeutic efficacy of inhibiting angiogenesis in this disease. Current therapeutic options for patients with metastatic hormone-refractory CaP are very limited. Targeting vasculature is a developing area, which shows promise for the control of late stage and recurrent CaP disease and for overcoming drug resistance. We discuss angiogenesis and its postulated mechanisms and focus on the regulation of angiogenesis in CaP progression and the therapeutic beneficial effects associated with targeting of the CaP vasculature to overcome the resistance to current treatments and CaP recurrence.
Collapse
Affiliation(s)
- Yong Li
- Cancer Care Centre, St George Hospital, Sydney, NSW, Australia.
| | | |
Collapse
|
132
|
Trerotola M, Rathore S, Goel HL, Li J, Alberti S, Piantelli M, Adams D, Jiang Z, Languino LR. CD133, Trop-2 and alpha2beta1 integrin surface receptors as markers of putative human prostate cancer stem cells. Am J Transl Res 2010; 2:135-144. [PMID: 20407603 PMCID: PMC2855629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Accepted: 03/10/2010] [Indexed: 05/29/2023]
Abstract
Cancer stem cells (CSCs) play a key role in initiation and development of cancer and are attractive targets for therapy. The identification of CSC surface receptors to be used as therapeutic targets in vivo remains a difficult task. In this study, we assessed the expression pattern of three surface receptors: CD133, Trop-2 and alpha2beta1 integrin in human prostate cancer in order to identify CSC-niches. CD133 was found to be expressed in small clusters of cells localized in focal areas of benign as well as malignant lesions, suggesting that this protein is a bona fide marker of the prostate stem/progenitor compartment. Trop-2 was localized in both basal and luminal layers of benign glands and was highly expressed in malignant lesions. Moreover, isolated cells in benign and malignant areas were found to co-express both CD133 and Trop-2. alpha2beta1 integrin was expressed in the prostatic epithelium as well as in the surrounding stroma, limiting its utility as a marker of CSCs. In summary, we demonstrate that the combination of CD133 and Trop-2 is useful to mark putative CSC-containing compartments in human prostate.
Collapse
|
133
|
Sakamoto S, Kyprianou N. Targeting anoikis resistance in prostate cancer metastasis. Mol Aspects Med 2010; 31:205-14. [PMID: 20153362 DOI: 10.1016/j.mam.2010.02.001] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Accepted: 02/06/2010] [Indexed: 01/20/2023]
Abstract
Anoikis is a mode of apoptotic cell death, consequential to insufficient cell-matrix interactions and a critical player in tumor angiogenesis and metastasis. The events involved in tumor cell progression toward metastasis potential are mediated by integrins, which upon engagement with components of the extracellular matrix (ECM), reorganize to form adhesion complexes. Targeting apoptotic players is of immense therapeutic significance since resistance to apoptosis is not only critical in conferring therapeutic failure to standard treatment strategies, but anoikis (apoptosis upon loss of anchorage and detachment from ECM) also plays an important role in angiogenesis and metastasis. The ability to survive in the absence of adhesion to the ECM, enables tumor cells to disseminate from the primary tumor site, invade a distant site and establish a metastatic lesion. Tumor cells can escape from detachment-induced apoptosis by controlling anoikis pathways, including the extrinsic death receptor pathway and the ECM-integrin mediated cell survival pathway. Considering the functional promiscuity of individual signaling effectors, it is critical to dissect the molecular networks mechanistically driving tumor cells to evade anoikis and embark on a metastatic spread. Resistance to die via anoikis dictates tumor cell survival and provides a molecular basis for therapeutic targeting of metastatic prostate cancer. Further dissection of critical anoikis signaling events will enable the therapeutic optimization of anoikis targeting to impair prostate cancer metastasis prior to its initiation. This review will discuss the molecular understanding of anoikis regulation in the tumor microenvironment and the in vivo pharmacological implementation of a novel class of antitumor-drugs to optimize apoptotic-based therapeutic targeting, bypassing anoikis-resistance to impair prostate cancer progression to metastasis. Potential combination strategies targeting tumor vascularity (via anoikis) and impairing tumor initiation (via "classic" apoptosis), provide strong therapeutic promise for metastatic prostate cancer by preventing the onset of metastasis.
Collapse
Affiliation(s)
- Shinichi Sakamoto
- Department of Surgery/Urology, University of Kentucky College of Medicine, Lexington, KY, USA
| | | |
Collapse
|
134
|
Soumetz FC, Saenz JF, Pastorino L, Ruggiero C, Nosi D, Raiteri R. Investigation of integrin expression on the surface of osteoblast-like cells by atomic force microscopy. Ultramicroscopy 2010; 110:330-8. [PMID: 20149538 DOI: 10.1016/j.ultramic.2010.01.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 11/27/2009] [Accepted: 01/12/2010] [Indexed: 11/25/2022]
Abstract
The transforming growth factor beta1 (TGF-beta1) is a human cytokine which has been demonstrated to modulate cell surface integrin repertoire. In this work integrin expression in response to TGF-beta1 stimulation has been investigated on the surface of human osteoblast-like cells. We used atomic force microscopy (AFM) and confocal laser scanning microscopy to assess integrin expression and to evaluate their distribution over the dorsal side of the plasma membrane. AFM probes have been covalently functionalized with monoclonal antibodies specific to the beta1 integrin subunit. Force curves have been collected in order to obtain maps of the interaction between the immobilized antibody and the respective cell membrane receptors. Adhesion peaks have been automatically detected by means of an ad hoc developed data analysis software. The specificity of the detected interactions has been assessed by adding free antibody in the solution and monitoring the dramatic decrease in the recorded interactions. In addition, the effect of TGF-beta1 treatment on both the fluorescence signal and the adhesion events has been tested. The level of expression of the beta1 integrin subunit was enhanced by TGF-beta1. As a further analysis, the adhesion force of the single living cells to the substrate was measured by laterally pushing the cell with the AFM tip and measuring the force necessary to displace it. The treatment with TGF-beta1 resulted in a decrease of the cell/substrate adhesion force. Results obtained by AFM have been validated by confocal laser scanning microscopy thus demonstrating the high potential of the AFM technique for the investigation of cell surface receptors distribution and trafficking at the nanoscale.
Collapse
Affiliation(s)
- Federico Caneva Soumetz
- Department of Communication, Computer and System Sciences, University of Genova, Via Opera Pia, 13-16145 Genova, Italy
| | | | | | | | | | | |
Collapse
|
135
|
Altieri DC, Languino LR, Lian JB, Stein JL, Leav I, van Wijnen AJ, Jiang Z, Stein GS. Prostate cancer regulatory networks. J Cell Biochem 2009; 107:845-52. [PMID: 19492418 DOI: 10.1002/jcb.22162] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Although the timing with which common epithelial malignancies arise and become established remains a matter of debate, it is clear that by the time they are detected these tumors harbor hundreds of deregulated, aberrantly expressed or mutated genes. This enormous complexity poses formidable challenges to identify gene pathways that are drivers of tumorigenesis, potentially suitable for therapeutic intervention. An alternative approach is to consider cancer pathways as interconnected networks, and search for potential nodal proteins capable of connecting multiple signaling networks of tumor maintenance. We have modeled this approach in advanced prostate cancer, a condition with current limited therapeutic options. We propose that the integration of three signaling networks, including chaperone-mediated mitochondrial homeostasis, integrin-dependent cell signaling, and Runx2-regulated gene expression in the metastatic bone microenvironment plays a critical role in prostate cancer maintenance, and offers novel options for molecular therapy.
Collapse
Affiliation(s)
- Dario C Altieri
- Department of Cancer Biology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA.
| | | | | | | | | | | | | | | |
Collapse
|
136
|
Gorlov IP, Byun J, Gorlova OY, Aparicio AM, Efstathiou E, Logothetis CJ. Candidate pathways and genes for prostate cancer: a meta-analysis of gene expression data. BMC Med Genomics 2009; 2:48. [PMID: 19653896 PMCID: PMC2731785 DOI: 10.1186/1755-8794-2-48] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Accepted: 08/04/2009] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The genetic mechanisms of prostate tumorigenesis remain poorly understood, but with the advent of gene expression array capabilities, we can now produce a large amount of data that can be used to explore the molecular and genetic mechanisms of prostate tumorigenesis. METHODS We conducted a meta-analysis of gene expression data from 18 gene array datasets targeting transition from normal to localized prostate cancer and from localized to metastatic prostate cancer. We functionally annotated the top 500 differentially expressed genes and identified several candidate pathways associated with prostate tumorigeneses. RESULTS We found the top differentially expressed genes to be clustered in pathways involving integrin-based cell adhesion: integrin signaling, the actin cytoskeleton, cell death, and cell motility pathways. We also found integrins themselves to be downregulated in the transition from normal prostate tissue to primary localized prostate cancer. Based on the results of this study, we developed a collagen hypothesis of prostate tumorigenesis. According to this hypothesis, the initiating event in prostate tumorigenesis is the age-related decrease in the expression of collagen genes and other genes encoding integrin ligands. This concomitant depletion of integrin ligands leads to the accumulation of ligandless integrin and activation of integrin-associated cell death. To escape integrin-associated death, cells suppress the expression of integrins, which in turn alters the actin cytoskeleton, elevates cell motility and proliferation, and disorganizes prostate histology, contributing to the histologic progression of prostate cancer and its increased metastasizing potential. CONCLUSION The results of this study suggest that prostate tumor progression is associated with the suppression of integrin-based cell adhesion. Suppression of integrin expression driven by integrin-mediated cell death leads to increased cell proliferation and motility and increased tumor malignancy.
Collapse
Affiliation(s)
- Ivan P Gorlov
- Department of Genitourinary Medical Oncology, The University of Texas M, D, Anderson Cancer Center, Houston, TX, USA.
| | | | | | | | | | | |
Collapse
|
137
|
Gorlov IP, Gallick GE, Gorlova OY, Amos C, Logothetis CJ. GWAS meets microarray: are the results of genome-wide association studies and gene-expression profiling consistent? Prostate cancer as an example. PLoS One 2009; 4:e6511. [PMID: 19652704 PMCID: PMC2714961 DOI: 10.1371/journal.pone.0006511] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Accepted: 06/29/2009] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWASs) and global profiling of gene expression (microarrays) are two major technological breakthroughs that allow hypothesis-free identification of candidate genes associated with tumorigenesis. It is not obvious whether there is a consistency between the candidate genes identified by GWAS (GWAS genes) and those identified by profiling gene expression (microarray genes). METHODOLOGY/PRINCIPAL FINDINGS We used the Cancer Genetic Markers Susceptibility database to retrieve single nucleotide polymorphisms from candidate genes for prostate cancer. In addition, we conducted a large meta-analysis of gene expression data in normal prostate and prostate tumor tissue. We identified 13,905 genes that were interrogated by both GWASs and microarrays. On the basis of P values from GWASs, we selected 1,649 most significantly associated genes for functional annotation by the Database for Annotation, Visualization and Integrated Discovery. We also conducted functional annotation analysis using same number of the top genes identified in the meta-analysis of the gene expression data. We found that genes involved in cell adhesion were overrepresented among both the GWAS and microarray genes. CONCLUSIONS/SIGNIFICANCE We conclude that the results of these analyses suggest that combining GWAS and microarray data would be a more effective approach than analyzing individual datasets and can help to refine the identification of candidate genes and functions associated with tumor development.
Collapse
Affiliation(s)
- Ivan P Gorlov
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | | | | | | |
Collapse
|
138
|
Goel HL, Moro L, Murphy-Ullrich JE, Hsieh CC, Wu CL, Jiang Z, Languino LR. Beta1 integrin cytoplasmic variants differentially regulate expression of the antiangiogenic extracellular matrix protein thrombospondin 1. Cancer Res 2009; 69:5374-82. [PMID: 19549894 DOI: 10.1158/0008-5472.can-09-0186] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Beta(1) integrins play an important role in regulating cell proliferation and survival. Using small interfering RNA or an inhibitory antibody to beta(1), we show here that, in vivo, beta(1) integrins are essential for prostate cancer growth. Among the five known beta(1) integrin cytoplasmic variants, two have been shown to differentially affect prostate cell functions. The beta(1A) variant promotes normal and cancer cell proliferation, whereas the beta(1C) variant, which is down-regulated in prostate cancer, inhibits tumor growth and appears to have a dominant effect on beta(1A). To investigate the mechanism by which beta(1C) inhibits the tumorigenic potential of beta(1A), we analyzed changes in gene expression in cells transfected with either beta(1C) or beta(1A). The results show that beta(1C) expression increases the levels of an extracellular matrix protein, thrombospondin 1 (TSP1), an angiogenesis inhibitor. TSP1 protein levels are increased upon beta(1C) expression in prostate cancer cells as well as in beta(1)-null GD25 cells. We show that TSP1 does not affect proliferation, apoptosis, or anchorage-independent growth of prostate cancer cells. In contrast, the newly synthesized TSP1, secreted by prostate cancer cells expressing beta(1C), prevents proliferation of endothelial cells. In conclusion, our novel findings indicate that expression of the beta(1C) integrin variant in prostate glands prevents cancer progression by up-regulation of TSP1 levels and inhibition of angiogenesis.
Collapse
Affiliation(s)
- Hira Lal Goel
- Department of Cancer Biology, Prostate Cancer Discovery and Development Program, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | | | | | | | | | |
Collapse
|
139
|
Feng X. Chemical and Biochemical Basis of Cell-Bone Matrix Interaction in Health and Disease. ACTA ACUST UNITED AC 2009; 3:189-196. [PMID: 20161446 DOI: 10.2174/187231309788166398] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Bone, a calcified tissue composed of 60% inorganic component (hydroxyapatite), 10% water and 30% organic component (proteins), has three functions: providing mechanical support for locomotion, protecting vital organs, and regulating mineral homeostasis. A lifelong execution of these functions depends on a healthy skeleton, which is maintained by constant bone remodeling in which old bone is removed by the bone-resorbing cell, osteoclasts, and then replaced by new bone formed by the bone-forming cell, osteoblasts. This remodeling process requires a physical interaction of bone with these bone cells. Moreover, numerous cancers including breast and prostate have a high tendency to metastasize to bone, which is in part attributable to the capacity of the tumor cells to attach to bone. The intensive investigation in the past two decades has led to the notion that the cell-bone interaction involves integrins on cell surface and bone matrix proteins. However, the biochemical composition of bone and emerging evidence are inconsistent with this belief. In this review, I will discuss the current understanding of the molecular mechanism underlying the cell-bone interaction. I will also highlight the facts and new findings supporting that the inorganic, rather than the organic, component of bone is likely responsible for cellular attachment.
Collapse
Affiliation(s)
- Xu Feng
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
140
|
Goel HL, Alam N, Johnson INS, Languino LR. Integrin signaling aberrations in prostate cancer. Am J Transl Res 2009; 1:211-220. [PMID: 19956432 PMCID: PMC2757165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 04/17/2009] [Indexed: 05/28/2023]
Abstract
Integrins are cell surface receptors for extracellular matrix proteins and play a key role in cell survival, proliferation, migration and gene expression. Integrin signaling has been shown to be deregulated in several types of cancer, including prostate cancer. This review is focused on integrin signaling pathways known to be deregulated in prostate cancer and known to promote prostate cancer progression.
Collapse
Affiliation(s)
- Hira Lal Goel
- Department of Cancer Biology, Prostate Cancer Discovery and Development Program, Cancer Center, University of Massachusetts Medical School Worcester, MA 01605, USA
| | | | | | | |
Collapse
|