1
|
Lengyel M, Molnár Á, Nagy T, Jdeed S, Garai I, Horváth Z, Uray IP. Zymogen granule protein 16B (ZG16B) is a druggable epigenetic target to modulate the mammary extracellular matrix. Cancer Sci 2025; 116:81-94. [PMID: 39489500 PMCID: PMC11711063 DOI: 10.1111/cas.16382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/19/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024] Open
Abstract
High tissue density of the mammary gland is considered a pro-tumorigenic factor, hence suppressing the stimuli that induce matrix buildup carries the potential for cancer interception. We found that in non-malignant mammary epithelial cells the combination of the chemopreventive agents bexarotene (Bex) and carvedilol (Carv) suppresses the zymogen granule protein 16B (ZG16B, PAUF) through an interaction of ARID1A with a proximal enhancer. Bex + Carv also reduced ZG16B levels in vivo in normal breast tissue and MDA-MB231 tumor xenografts. The relevance of ZG16B is underscored by ongoing clinical trials targeting ZG16B in pancreatic cancers, but its role in breast cancer development is unclear. In immortalized mammary epithelial cells, secreted recombinant ZG16B stimulated mitogenic kinase phosphorylation, detachment and mesenchymal characteristics, and promoted proliferation, motility and clonogenic growth. Highly concerted induction of specific laminin, collagen and integrin isoforms indicated a shift in matrix properties toward increased density and cell-matrix interactions. Exogenous ZG16B alone blocked Bex + Carv-mediated control of cell growth and migration, and antagonized Bex + Carv-induced gene programs regulating cell adhesion and migration. In breast cancer cells ZG16B induced colony formation and anchorage-independent growth, and stimulated migration in a PI3K/Akt-dependent manner. In contrast, Bex + Carv inhibited colony formation, reduced Ki67 levels, ZG16B expression and glucose uptake in MDA-MB231 xenografts. These data establish ZG16B as a druggable pro-tumorigenic target in breast cell transformation and suggest a key role of the matrisome network in rexinoid-dependent antitumor activity.
Collapse
Affiliation(s)
- Máté Lengyel
- Department of Clinical Oncology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- The Molecular Cell and Immune Biology Doctoral SchoolUniversity of DebrecenDebrecenHungary
| | - Ádám Molnár
- Department of Clinical Oncology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Tamás Nagy
- Department of Nuclear MedicineUniversity of DebrecenDebrecenHungary
| | - Sham Jdeed
- Department of Clinical Oncology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- The Molecular Cell and Immune Biology Doctoral SchoolUniversity of DebrecenDebrecenHungary
| | - Ildikó Garai
- Department of Nuclear MedicineUniversity of DebrecenDebrecenHungary
| | - Zsolt Horváth
- Center of OncoradiologyBács‐Kiskun County Teaching HospitalKecskemétHungary
| | - Iván P. Uray
- Department of Clinical Oncology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- The Molecular Cell and Immune Biology Doctoral SchoolUniversity of DebrecenDebrecenHungary
- Department of Biochemistry and Molecular BiologyUniversity of DebrecenDebrecenHungary
| |
Collapse
|
2
|
Venom peptides in cancer therapy: An updated review on cellular and molecular aspects. Pharmacol Res 2020; 164:105327. [PMID: 33276098 DOI: 10.1016/j.phrs.2020.105327] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023]
Abstract
Based on the high incidence and mortality rates of cancer, its therapy remains one of the most vital challenges in the field of medicine. Consequently, enhancing the efficacy of currently applied treatments and finding novel strategies are of great importance for cancer treatment. Venoms are important sources of a variety of bioactive compounds including salts, small molecules, macromolecules, proteins, and peptides that are defined as toxins. They can exhibit different pharmacological effects, and in recent years, their anti-tumor activities have gained significant attention. Several different compounds are responsible for the anti-tumor activity of venoms, and peptides are one of them. In the present review, we discuss the possible anti-tumor activities of venom peptides by highlighting molecular pathways and mechanisms through which these molecules can act effectively. Venom peptides can induce cell death in cancer cells and can substantially enhance the efficacy of chemotherapy and radiotherapy. Also, the venom peptides can mitigate the migration of cancer cells via suppression of angiogenesis and epithelial-to-mesenchymal transition. Notably, nanoparticles have been applied in enhancing the bioavailability of venom peptides and providing targeted delivery, thereby leading to their elevated anti-tumor activity and potential application for cancer therapy.
Collapse
|
3
|
Kummer D, Steinbacher T, Schwietzer MF, Thölmann S, Ebnet K. Tetraspanins: integrating cell surface receptors to functional microdomains in homeostasis and disease. Med Microbiol Immunol 2020; 209:397-405. [PMID: 32274581 PMCID: PMC7395057 DOI: 10.1007/s00430-020-00673-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/28/2020] [Indexed: 12/27/2022]
Abstract
Tetraspanins comprise a family of proteins embedded in the membrane through four transmembrane domains. One of the most distinctive features of tetraspanins is their ability to interact with other proteins in the membrane using their extracellular, transmembrane and cytoplasmic domains, allowing them to incorporate several proteins into clusters called tetraspanin-enriched microdomains. The spatial proximity of signaling proteins and their regulators enables a rapid functional cross-talk between these proteins, which is required for a rapid translation of extracellular signals into intracellular signaling cascades. In this article, we highlight a few examples that illustrate how tetraspanin-mediated interactions between cell surface proteins allow their functional cross-talk to regulate intracellular signaling.
Collapse
Affiliation(s)
- Daniel Kummer
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
- Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany
| | - Tim Steinbacher
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Mariel Flavia Schwietzer
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Sonja Thölmann
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Klaus Ebnet
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany.
- Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany.
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany.
- Institute of Medical Biochemistry, ZMBE, Von-Esmarch-Str. 56, 48149, Münster, Germany.
| |
Collapse
|
4
|
Margetts PJ, Bonniaud P. Basic Mechanisms and Clinical Implications of Peritoneal Fibrosis. Perit Dial Int 2020. [DOI: 10.1177/089686080302300604] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Peter J. Margetts
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Philippe Bonniaud
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
5
|
Isolation of an Anti-Tumour Disintegrin: Dabmaurin-1, a Peptide Lebein-1-Like, from Daboia mauritanica Venom. Toxins (Basel) 2020; 12:toxins12020102. [PMID: 32033352 PMCID: PMC7076848 DOI: 10.3390/toxins12020102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/28/2020] [Accepted: 01/31/2020] [Indexed: 11/17/2022] Open
Abstract
In the soft treatment of cancer tumours, consequent downregulation of the malignant tissue angiogenesis constitutes an efficient way to stifle tumour development and metastasis spreading. As angiogenesis requires integrin–promoting endothelial cell adhesion, migration, and vessel tube formation, integrins represent potential targets of new therapeutic anti–angiogenic agents. Our work is a contribution to the research of such therapeutic disintegrins in animal venoms. We report isolation of one peptide, named Dabmaurin–1, from the hemotoxic venom of snake Daboia mauritanica, and we evaluate its potential anti–tumour activity through in vitro inhibition of the human vascular endothelial cell HMECs functions involved in tumour angiogenesis. Dabmaurin–1 altered, in a dose–dependent manner, without any significant cytotoxicity, HMEC proliferation, adhesion, and their mesenchymal migration onto various extracellular matrix proteins, as well as formation of capillary–tube mimics on MatrigelTM. Via experiments involving HMEC or specific cancers cells integrins, we demonstrated that the above Dabmaurin–1 effects are possibly due to some anti–integrin properties. Dabmaurin–1 was demonstrated to recognize a broad panel of prooncogenic integrins (αvβ6, αvβ3 or αvβ5) and/or particularly involved in control of angiogenesis (α5β1, α6β4, αvβ3 or αvβ5). Furthermore, mass spectrometry and partial N–terminal sequencing of this peptide revealed, it is close to Lebein–1, a known anti–β1 disintegrin from Macrovipera lebetina venom. Therefore, our results show that if Dabmaurin–1 exhibits in vitro apparent anti–angiogenic effects at concentrations lower than 30 nM, it is likely because it acts as an anti–tumour disintegrin.
Collapse
|
6
|
Gong L, Zheng Y, Liu S, Peng Z. Fibronectin Regulates the Dynamic Formation of Ovarian Cancer Multicellular Aggregates and the Expression of Integrin Receptors. Asian Pac J Cancer Prev 2018; 19:2493-2498. [PMID: 30256042 PMCID: PMC6249468 DOI: 10.22034/apjcp.2018.19.9.2493] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: To investigate the regulatory role of fibronectin (FN) in the formation of multicellular aggregate (MCA) in ovarian cancer SKOV3 and OVCAR-3 cells and integrin expression. Methods: The dynamic formation of MCA in SKOV3 and OVCAR-3 was determined using the liquid overlay technique in the presence or absence of FN, anti-FN, RGD peptide, control RGE. The expression of α3β1, α4β1 and α5β1 integrin in monolayer cells, MCA and FN-treated MCA were determined by flow cytometry and quantitative RT-PCR. Results: OVCAR-3 and SKOV3 MCA were formed on the 4th and 8th day and peaked on the 6th and 9th day, respectively. Treatment with different concentrations of FN, LN, type IV collagen and control RGE peptide promoted MCA growth, which was mitigated by anti-FN and RGD peptide. In comparison with monolayer cells, up-regulated α3β1, α4β1 and α5β1 expression were detected in MCA while treatment with FN in both cells. Conclusions: OVCAR-3 and SKOV3 cells had varying dynamic formation of MCA in our experimental system. FN enhanced MCA formation in both cells, which was associated with increased expression of 3β1, α4β1 and α5β1 in the MCA. Therefore, FN and these integrins may be new therapeutic targets for intervention of ovarian cancer metastasis.
Collapse
Affiliation(s)
- Lin Gong
- Department of Obstetrics and Gynecology, West China Second Hospital, West China Center of Medical Sciences, Sichuan University,Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children ,Sichuan University, Chengdu, China.
| | | | | | | |
Collapse
|
7
|
Kakei Y, Teraoka S, Akashi M, Hasegawa T, Komori T. Changes in cell junctions induced by inhibition of epidermal growth factor receptor in oral squamous cell carcinoma cells. Exp Ther Med 2017; 14:953-960. [PMID: 28810546 PMCID: PMC5525654 DOI: 10.3892/etm.2017.4606] [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: 11/09/2016] [Accepted: 03/17/2017] [Indexed: 11/29/2022] Open
Abstract
The benefits of epidermal growth factor receptor (EGFR) targeting in the treatment of head and neck cancer, have been documented. However, a minority of patients with head and neck cancer are unresponsive to EGFR targeting therapies. The present study evaluated the effects and limitations of an EGFR inhibitor on oral squamous cell carcinoma cells, particularly on cell-cell junctions mediated by epithelial (E)-cadherin. HSC-3 oral squamous cell carcinoma cells were treated with the EGFR inhibitor, AG1478 (0, 0.5, 2, 10 and 50 µM), and the effects of EGFR inhibition in HSC-3 cells were evaluated by wound healing assays, E-cadherin immunostaining and measurement of transepithelial electrical resistance in vitro. It was observed that treatment of oral squamous cell carcinoma cells with AG1478 suppressed cell motility, altered cell morphology and increased the number of cell-cell junctions compared with untreated control cells. Knockdown of EGFR induced a similar phenotype to that observed by the inhibition of EGFR. Furthermore, in oral squamous cell carcinoma cells treated with high-dose EGFR inhibitor (50 µM), the small number of cells that survived formed cell-cell junctions that were positive for E-cadherin expression. In cells treated with low concentrations of EGFR inhibitor (2 µM), recovery of epithelial properties was observed. The retention of E-cadherin expression in cells that survived high-dose EGFR inhibitor treatment may be a survival mechanism of cancer cells.
Collapse
Affiliation(s)
- Yasumasa Kakei
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Hyōgo 650-0017, Japan
| | - Shun Teraoka
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Hyōgo 650-0017, Japan
| | - Masaya Akashi
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Hyōgo 650-0017, Japan
| | - Takumi Hasegawa
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Hyōgo 650-0017, Japan
| | - Takahide Komori
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Hyōgo 650-0017, Japan
| |
Collapse
|
8
|
Li S, Li J, Hu T, Zhang C, Lv X, He S, Yan H, Tan Y, Wen M, Lei M, Zuo J. Bcl-2 overexpression contributes to laryngeal carcinoma cell survival by forming a complex with Hsp90β. Oncol Rep 2016; 37:849-856. [PMID: 27959448 DOI: 10.3892/or.2016.5295] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/19/2016] [Indexed: 11/06/2022] Open
Abstract
Laryngeal carcinoma (LC) is one of the most common malignant tumors of all head and neck squamous cell carcinomas (HNSCCs). However, the molecular mechanism and genetic basis of the development of LC have not been fully elucidated. To explore the possible mechanism, targeted proteomic analysis was performed on Bcl-2-associated proteins from LC cells. According to our results, 35 proteins associated with Bcl-2 were identified and Hsp90β was confirmed by co-immunoprecipitation and western blot analysis. Protein‑protein interaction (PPI) analysis indicated that Bcl-2‑Hsp90β interactions may be involved in the anti-apoptotic progression of LC. Further results revealed that disruption of the Bcl-2-Hsp90β interaction inhibited the anti-apoptotic ability of Bcl-2 and decreased the caspase activation in LC, which has broad implications for the better understanding of tumor formation, tumor cell survival and development of metastasis due to Bcl-2. Collectively, we report the mechanism by which Bcl-2 functions in LC as an anti-apoptotic factor in relation to its association with proteins and potentially identify a Bcl-2/Hsp90β axis as a novel target for LC therapy.
Collapse
Affiliation(s)
- Sai Li
- Oncology Department, The Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jincheng Li
- Medical School, Shaoyang University, Shaoyang, Hunan 422000, P.R. China
| | - Tian Hu
- Oncology Department, The Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Chuhong Zhang
- Oncology Department, The Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiu Lv
- Oncology Department, The Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Sha He
- Oncology Department, The Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hanxing Yan
- Oncology Department, The Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yixi Tan
- Oncology Department, The Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Meiling Wen
- Oncology Department, The Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Mingsheng Lei
- Department of Respiratory and Critical Care Medicine, The People's Hospital of Zhangjiajie City, Zhangjiajie, Hunan 427000, P.R. China
| | - Jianhong Zuo
- Oncology Department, The Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
9
|
Caldeira J, Figueiredo J, Brás-Pereira C, Carneiro P, Moreira AM, Pinto MT, Relvas JB, Carneiro F, Barbosa M, Casares F, Janody F, Seruca R. E-cadherin-defective gastric cancer cells depend on Laminin to survive and invade. Hum Mol Genet 2015; 24:5891-900. [PMID: 26246502 DOI: 10.1093/hmg/ddv312] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/29/2015] [Indexed: 01/05/2023] Open
Abstract
Epithelial-cadherin (Ecad) deregulation affects cell-cell adhesion and results in increased invasiveness of distinct human carcinomas. In gastric cancer, loss of Ecad expression is a common event and is associated with disease aggressiveness and poor prognosis. However, the molecular mechanisms underlying the invasive process associated to Ecad dysfunction are far from understood. We hypothesized that deregulation of cell-matrix interactions could play an important role during this process. Thus, we focussed on LM-332, which is a major matrix component, and in Ecad/LM-332 crosstalk in the process of Ecad-dependent invasion. To verify whether matrix deregulation was triggered by Ecad loss, we used the Drosophila model. To dissect the key molecules involved and unveil their functional significance, we used gastric cancer cell lines. The relevance of this relationship was then confirmed in human primary tumours. In vivo, Ecad knockdown induced apoptosis; nonetheless, at the invasive front, cells ectopically expressed Laminin A and βPS integrin. In vitro, we demonstrated that, in two different gastric cancer cell models, Ecad-defective cells overexpressed Laminin γ2 (LM-γ2), β1 and β4 integrin, when compared with Ecad-competent ones. We showed that LM-γ2 silencing impaired invasion and enhanced cell death, most likely via pSrc and pAkt reduction, and JNK activation. In human gastric carcinomas, we found a concomitant decrease in Ecad and increase in LM-γ2. This is the first evidence that ectopic Laminin expression depends on Ecad loss and allows Ecad-dysfunctional cells to survive and invade. This opens new avenues for using LM-γ2 signalling regulators as molecular targets to impair gastric cancer progression.
Collapse
Affiliation(s)
- Joana Caldeira
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal, Andalusian Centre for Developmental Biology (CABD), Seville, Spain, Instituto de Engenharia Biomédica (INEB), Instituto de Investigação e Inovação em Saúde (i3S)
| | - Joana Figueiredo
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal, Instituto de Investigação e Inovação em Saúde (i3S)
| | | | - Patrícia Carneiro
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal, Instituto de Investigação e Inovação em Saúde (i3S)
| | - Ana M Moreira
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal, Instituto de Investigação e Inovação em Saúde (i3S)
| | - Marta T Pinto
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal, Instituto de Investigação e Inovação em Saúde (i3S)
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde (i3S), Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Fátima Carneiro
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal, Instituto de Investigação e Inovação em Saúde (i3S), Department of Pathology and Oncology, Medical Faculty of the University of Porto, Porto, Portugal, Centro Hospitalar São João, Porto, Portugal and
| | - Mário Barbosa
- Instituto de Engenharia Biomédica (INEB), Instituto de Investigação e Inovação em Saúde (i3S), Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto (ICBAS), Porto, Portugal
| | - Fernando Casares
- Andalusian Centre for Developmental Biology (CABD), Seville, Spain
| | | | - Raquel Seruca
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal, Instituto de Investigação e Inovação em Saúde (i3S), Department of Pathology and Oncology, Medical Faculty of the University of Porto, Porto, Portugal,
| |
Collapse
|
10
|
Zhao H, Yang F, Zhao W, Zhang C, Liu J. Fascin Overexpression Promotes Cholangiocarcinoma RBE Cell Proliferation, Migration, and Invasion. Technol Cancer Res Treat 2015; 15:322-33. [PMID: 25882880 DOI: 10.1177/1533034615580696] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/28/2015] [Indexed: 01/04/2023] Open
Abstract
Fascin is overexpressed in various tumor tissues and is closely related to tumor metastasis and invasion. However, the role of fascin in cholangiocarcinoma RBE cells has not been clearly reported. This study aimed to establish a cholangiocarcinoma cell line with stable and high expression of fascin to observe the effect of fascin on cell proliferation, migration, and invasion. A fascin overexpression vector, pcDNA3.1-Fascin, was constructed and transfected into the human cholangiocarcinoma RBE cell line. The results of real-time polymerase chain reaction, Western blot, and immunofluorescence indicated that fascin was steadily and highly expressed in RBE cells. The results of 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide and colony formation assay indicated that upregulated fascin expression could enhance cholangiocarcinoma cell proliferation. The results of wound healing assay and transwell assay indicated that fascin could promote cholangiocarcinoma cell migration and invasion, and a further study found that the nuclear factor-κB signaling pathway was activated after upregulation of fascin, whereas E-cadherin expression in these cells was significantly decreased. Additionally, E-cadherin expression was significantly increased after inhibiting nuclear factor-κB activity using inhibitor or small interfering RNA, and E-cadherin expression was decreased by fascin overexpression after nuclear factor-κB inhibition, suggesting that nuclear factor-κB signaling pathway was not involved in the regulation of E-cadherin by fascin. In summary, the results of this study demonstrated that fascin effectively promoted cholangiocarcinoma RBE cell proliferation, migration, and invasion. This study provides evidence for fascin as a potential target in the treatment of cholangiocarcinoma.
Collapse
Affiliation(s)
- Haiying Zhao
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Fuquan Yang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wenyan Zhao
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chunjv Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jingang Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Bertram K, Valcu CM, Weitnauer M, Linne U, Görlach A. NOX1 supports the metabolic remodeling of HepG2 cells. PLoS One 2015; 10:e0122002. [PMID: 25806803 PMCID: PMC4373763 DOI: 10.1371/journal.pone.0122002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/09/2015] [Indexed: 12/31/2022] Open
Abstract
NADPH oxidases are important sources of reactive oxygen species (ROS) which act as signaling molecules in the regulation of protein expression, cell proliferation, differentiation, migration and cell death. The NOX1 subunit is over-expressed in several cancers and NOX1 derived ROS have been repeatedly linked with tumorigenesis and tumor progression although underlying pathways are ill defined. We engineered NOX1-depleted HepG2 hepatoblastoma cells and employed differential display 2DE experiments in order to investigate changes in NOX1-dependent protein expression profiles. A total of 17 protein functions were identified to be dysregulated in NOX1-depleted cells. The proteomic results support a connection between NOX1 and the Warburg effect and a role for NOX in the regulation of glucose and glutamine metabolism as well as of lipid, protein and nucleotide synthesis in hepatic tumor cells. Metabolic remodeling is a common feature of tumor cells and understanding the underlying mechanisms is essential for the development of new cancer treatments. Our results reveal a manifold involvement of NOX1 in the metabolic remodeling of hepatoblastoma cells towards a sustained production of building blocks required to maintain a high proliferative rate, thus rendering NOX1 a potential target for cancer therapy.
Collapse
Affiliation(s)
- Katharina Bertram
- Experimental and Molecular Paediatric Cardiology, German Heart Centre Munich at the Technical University Munich, Lazarettstr. 36, Munich, Germany
| | - Cristina-Maria Valcu
- Experimental and Molecular Paediatric Cardiology, German Heart Centre Munich at the Technical University Munich, Lazarettstr. 36, Munich, Germany
- * E-mail: (CMV), (AG)
| | - Michael Weitnauer
- Experimental and Molecular Paediatric Cardiology, German Heart Centre Munich at the Technical University Munich, Lazarettstr. 36, Munich, Germany
| | - Uwe Linne
- Chemistry Department—Mass Spectrometry, Philipps-University Marburg, Hans-Meerwein-Strasse, Marburg, Germany
| | - Agnes Görlach
- Experimental and Molecular Paediatric Cardiology, German Heart Centre Munich at the Technical University Munich, Lazarettstr. 36, Munich, Germany
- * E-mail: (CMV), (AG)
| |
Collapse
|
12
|
Immunoexpression of α2β1, α3β1, and α5β1 integrins in pleomorphic adenoma and adenoid cystic carcinoma. Appl Immunohistochem Mol Morphol 2012; 21:258-64. [PMID: 22914615 DOI: 10.1097/pai.0b013e3182649119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The aim of the present study was to compare the expression of α2β1, α3β1, and α5β1 integrins between 28 pleomorphic adenomas (PAs) and 10 adenoid cystic carcinomas (ACCs), and investigate differences in the expression of these integrins according to histologic subtypes of ACCs. It was taken into consideration the presence or absence, distribution, and localization of integrin immunoexpression. There was immunoreactivity in the intercellular contacts of the strands, nests, and solid sheets of PAs, as well as in the luminal and nonluminal cells of the duct-like structures, with a predominant immunoexpression in the luminal cells. The immunoexpression in ACCs varied with histologic subtype of the tumor. It was verified for a tendency of absence and/or reduced expression of all integrins in the solid subtype of ACCs. In general, PAs revealed a more diffuse and remarkable immunoexpression of all studied integrins than ACCs. The reduced integrins expression in ACC may be related to a lesser degree of cell differentiation in this neoplasm. Moreover, the absence and/or reduced expression of the studied integrins in solid ACC suggest a possible role in pathogenesis and more aggressive biological behavior of this histologic subtype.
Collapse
|
13
|
Zhou L, Wang DS. Fascin-1 and digestive system carcinomas. Shijie Huaren Xiaohua Zazhi 2012; 20:2125-2130. [DOI: 10.11569/wcjd.v20.i23.2125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Invasion and metastasis are major reasons for poor prognosis of digestive system carcinomas. Motility and migratory capacity are important in contributing to tumor cell invasion and metastasis. Fascin-1 is a globular actin crosslinking protein that can form parallel actin bundles in cell protrusions and is involved in cell adhesion, movement, and signal transduction. In vitro up-regulation of Fascin-1 can increase migration and invasion capacity of cells, while down-regulation of Fascin-1 can decrease migration and invasion capacity of cells. Many studies show that up-regulation of Fascin-1 expression is significantly associated with worse prognosis, poorer differentiation, advanced TNM stage, lymph node metastasis, and distant metastasis in patients with digestive system carcinomas. Therefore, Fascin-1 may have prognostic value as an early biomarker for more aggressive digestive system carcinomas and may be a potential therapeutic target for tumor invasion and metastasis.
Collapse
|
14
|
Role of integrins in the carcinogenesis of squamous cell carcinoma of the tongue and lower lip. Appl Immunohistochem Mol Morphol 2012; 21:154-8. [PMID: 22595951 DOI: 10.1097/pai.0b013e31825905e5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE AND METHODS This study analyzed the distribution, intensity, and pattern of immunohistochemical expression of α2β1, α3β1, and α5β1 integrins in squamous cell carcinoma (SCC) of the lower lip and tongue to identify biomarkers that reflect the clinical course of this cancer. Immunoexpression was compared considering prognostic parameters such as anatomic site, metastasis, and histologic grade of malignancy. RESULTS Immunohistochemical analysis at the invasion front showed a predominance of granular cytoplasmic expression of the integrins studied. In most cases, immunopositive cells were diffusely distributed in the tumors, irrespective of their location, except for α3β1 integrin-positive cells which were focally distributed in 53.3% of tongue SCC cases. With respect to staining intensity, positive staining for α2β1 integrin was observed in 80% of lower lip SCCs and in 93.3% of tongue SCCs. Staining for α3β1 integrin was moderately positive in 60% of lower lip and tongue SCCs. The staining intensity of α5β1 integrin was moderately and strongly positive in 53.3% and 46.7% of lower lip SCCs, respectively, and in 46.7% and 53.3% of tongue SCCs. CONCLUSIONS The strong immunoreactivity for integrins α2β1, α3β1, and α5β1 seen in the oral SCC cases studied suggests a significant participation of these proteins in oral carcinogenesis. However, their expression does not reflect the clinical course of this cancer.
Collapse
|
15
|
Fernandez JM, Molinuevo MS, Cortizo AM, McCarthy AD, Cortizo MS. Characterization of poly(epsilon-caprolactone)/polyfumarate blends as scaffolds for bone tissue engineering. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2010; 21:1297-312. [PMID: 20534186 DOI: 10.1163/092050609x12517190417632] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
There is considerable interest in the design of polymeric biomaterials that can be used for the repair of bone defects. In this study, we used ultrasound to prepare a compatibilized blend of poly(epsilon-caprolactone) (PCL) and poly(diisopropyl fumarate) (PDIPF). The formation of post-sonication inter-polymer coupling products was verified by SEC analysis of a blend with azo-labeled PDIPF. We also analyzed the physicochemical and mechanical properties of the compatibilized blend. When compared to PCL alone, the PCL/PDIPF blend showed no difference in its resistance as evaluated by the elastic modulus, although it did show a 50% decrease in ultimate tensile stress (P < 0.05) and an 84% decrease in elongation-at-break (P < 0.05). However, the mechanical properties of this blend were comparable to those of trabecular bone. We next evaluated biocompatibility of the PCL/PDIPF blend, and of homo-polymeric PCL and PDIPF films for comparison, with UMR106 and MC3T3E1 osteoblastic cells. Osteoblasts plated on the compatibilized blend adhered and proliferated more than on either homo-polymer, showed a greater number of cellular processes with a better organized actin cytoskeleton and expressed more type-I collagen and mineral, both markers of osteoblast phenotype. These results support the hypothesis that this new compatibilized blend could be useful in future applications for bone regeneration.
Collapse
Affiliation(s)
- Juan Manuel Fernandez
- Instituto de Investigaciones Fisicoquímica Teóricas y Aplicadas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, CC 16 Suc. 4, 1900 La Plata, Argentina
| | | | | | | | | |
Collapse
|
16
|
Zuo J, Ishikawa T, Boutros S, Xiao Z, Humtsoe JO, Kramer RH. Bcl-2 Overexpression Induces a Partial Epithelial to Mesenchymal Transition and Promotes Squamous Carcinoma Cell Invasion and Metastasis. Mol Cancer Res 2010; 8:170-82. [PMID: 20145039 DOI: 10.1158/1541-7786.mcr-09-0354] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Jianhong Zuo
- Department of Cell and Tissue Biology, University of California San Francisco, 521 Parnassus Avenue, San Francisco, CA 94143-0640, USA
| | | | | | | | | | | |
Collapse
|
17
|
Ishikawa T, Kramer RH. Sdc1 negatively modulates carcinoma cell motility and invasion. Exp Cell Res 2009; 316:951-65. [PMID: 20036233 DOI: 10.1016/j.yexcr.2009.12.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Revised: 12/15/2009] [Accepted: 12/16/2009] [Indexed: 10/20/2022]
Abstract
During cancer progression, tumor cells eventually invade the surrounding collagen-rich extracellular matrix. Here we show that squamous cell carcinoma cells strongly adhere to Type I collagen substrates but display limited motility and invasion on collagen barriers. Further analysis revealed that in addition to the alpha2beta1 integrin, a second collagen receptor was identified as Syndecan-1 (Sdc1), a cell surface heparan sulfate proteoglycan. We demonstrate that siRNA-mediated depletion of Sdc1 reduced adhesion efficiency to collagen I, whereas knockdown of Sdc4 was without effect. Importantly, silencing Sdc1 expression caused reduced focal adhesion plaque formation and enhanced cell spreading and motility on collagen I substrates, but did not alter cell motility on other ECM substrates. Sdc1 depletion ablated adhesion-induced RhoA activation. In contrast, Rac1 was strongly activated following Sdc1 knockdown, suggesting that Sdc1 may mediate the link between integrin-induced actin remodeling and motility. Taken together, these data substantiate the existence of a co-adhesion receptor system in tumor cells, whereby Sdc1 functions as a key regulator of cell motility and cell invasion by modulating RhoA and Rac activity. Downregulation of Sdc1 expression during carcinoma progression may represent a mechanism by which tumor cells become more invasive and metastatic.
Collapse
Affiliation(s)
- Tohru Ishikawa
- Department of Cell and Tissue Biology, University of California San Francisco, 521 Parnassus Avenue, Room C-640, San Francisco, CA 94143-0640, USA
| | | |
Collapse
|
18
|
Humtsoe JO, Kramer RH. Differential epidermal growth factor receptor signaling regulates anchorage-independent growth by modulation of the PI3K/AKT pathway. Oncogene 2009; 29:1214-26. [PMID: 19935697 PMCID: PMC2829113 DOI: 10.1038/onc.2009.419] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Tumor cells are capable of surviving loss of nutrients and anchorage in hostile microenvironments. Under these conditions adapting to specific signaling pathways may shift the balance between growth and cellular dormancy. Here we report a mechanism by which EGFR differentially modulates the PI3K/AKT pathway in cellular stress conditions. When carcinoma cells were cultured as multicellular aggregates (MCA), cyclin D1 was induced through a serum-dependent EGFR activating pathway, triggering cell proliferation. The expression of cyclin D1 required both EGFR-mediated ERK and AKT activation. In serum-starved MCAs, EGFR activation was associated with active ERK1/2 but not AKT and failed to induce cyclin D1. Analysis revealed that, under serum-starved conditions, EGFR-Y1086 residue was poorly autophosphorylated and this correlated with failure to phosphorylate Gab1. Accordingly, the EGFR activation failed to induce EGFR/PI3K complex formation or AKT activation, preventing cyclin D1 induction. Furthermore, we show that in serum-starved MCA, expression of constitutively active AKT re-established cyclin D1 expression and induced proliferation in an EGFR-dependent manner. Thus, modulation of the PI3K/AKT pathway by context-dependent EGFR signaling may regulate tumor cell growth and dormancy.
Collapse
Affiliation(s)
- J O Humtsoe
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143-0640, USA
| | | |
Collapse
|
19
|
The response of endothelial cells to polymer surface composed of nanometric micelles. N Biotechnol 2009; 25:235-43. [PMID: 19429543 DOI: 10.1016/j.nbt.2009.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Revised: 01/08/2009] [Accepted: 01/08/2009] [Indexed: 11/22/2022]
Abstract
The response of endothelial cells (ECs) to a polyurethane (PU) characteristic of surface micelles ( approximately 89nm in diameter) and two novel nanocomposites (PU-Au) and (PU-Ag) containing smaller surface micelles ( approximately 14-22nm in diameter) was investigated. The molecular mechanism by which ECs reacted to nanometric surface micelles was examined. On PU-Au and PU-Ag, cell migration rate was promoted. This was accompanied by the upregulation of endothelial nitric oxide synthase (eNOS) and phosphorylated-Akt (p-Akt) expression. The induced eNOS and p-Akt expression was inhibited by LY294002, indicating that the effect of nanocomposites could be attributed to the PI3K pathway. An elevation of intracellular calcium concentration was noted. Additionally, more actin fibers were induced by PU-Au and PU-Ag. Reduction of actin expression upon addition of Y-27632 (an inhibitor of Rho-GTPase) and SU-1498 (an inhibitor of VEGF-R2) was observed. It was concluded that the nanometric micelles on PU surface may interact with ECs and accelerate their migration by increasing cytoplasmic Ca(2+) and stimulating the PI3K/Akt/eNOS signaling pathway.
Collapse
|
20
|
Ji Q, Burk RD. Downregulation of integrins by von Hippel-Lindau (VHL) tumor suppressor protein is independent of VHL-directed hypoxia-inducible factor alpha degradation. Biochem Cell Biol 2008; 86:227-34. [PMID: 18523483 DOI: 10.1139/o08-035] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene occurs in the majority of clear-cell renal cell carcinomas (RCCs). It was previously shown that VHL decreased the abundance of integrins alpha2, alpha5, and beta1, which is consistent with VHL-associated changes in cell-cell and cell - extracellular matrix adhesions. We investigated the mechanism by which VHL downregulates integrins. Although VHL can target hypoxia-inducible factor alpha (HIFalpha) subunits for degradation, VHL-dependent reduction of integrins was independent of O2 concentration and HIFalpha levels. VHL reduced the half-lives of integrins, and this activity was blocked by proteasomal inhibition. Although ectopically expressed FLAG-VHL retained HIFalpha degradation activity, it neither downregulated integrins nor promoted adherens and tight intercellular junctions, in contrast to expressed wild-type VHL. Moreover, integrins co-immunoprecipitated with wild-type VHL, but not FLAG-VHL. These data indicate that the downregulation of integrins by VHL is distinct from the regulation of HIFalpha subunits by VHL, and suggests that the loss of this activity contributes to VHL-associated RCC development through disruption of adherens and tight junctions.
Collapse
Affiliation(s)
- Qingzhou Ji
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
21
|
Bachy S, Letourneur F, Rousselle P. Syndecan-1 interaction with the LG4/5 domain in laminin-332 is essential for keratinocyte migration. J Cell Physiol 2008; 214:238-49. [PMID: 17579341 DOI: 10.1002/jcp.21184] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Laminin 5/laminin 332 (LN332) is an adhesion substrate for epithelial cells. After secretion of LN332, a regulated cleavage occurs at the carboxy-terminus of its alpha3 subunit, which releases a tandem of two globular modules named LG4/5. We show that the presence of the LG4/5 domain in precursor LN332 decreases its integrin-mediated cell adhesion properties in comparison with mature LN332. Whereas cell adhesion to the recombinant LG4/5 fragment relies solely on the heparan sulfate proteoglycan (HSPG) receptor syndecan-1, we reveal that both syndecan-1 and the alpha3beta1 integrin bind to precursor LN332. We further demonstrate that syndecan-1 mediated cell adhesion to the LG4/5 fragment and pre-LN332 allows the formation of fascin-containing protrusions, depending on the GTPases Rac and Cdc42 activation. Reducing syndecan-1 expression in normal keratinocytes prevents cell protrusions on pre-LN332 with subsequent failure of the peripheral localization of the alpha3beta1 integrin. We finally show that cell migration on pre-LN332 requires syndecan-1. Therefore, the LG4/5 domain in precursor LN332 appears to trigger intracellular signaling events, which participate in keratinocyte motility.
Collapse
Affiliation(s)
- Sophie Bachy
- IFR128 BioSciences Lyon-Gerland, Institut de Biologie et Chimie des Protéines, UMR 5086, CNRS, University Lyon1, 7 passage du Vercors, Lyon, France
| | | | | |
Collapse
|
22
|
Abstract
Forces are increasingly recognized as major regulators of cell structure and function, and the mechanical properties of cells are essential to the mechanisms by which cells sense forces, transmit them to the cell interior or to other cells, and transduce them into chemical signals that impact a spectrum of cellular responses. Comparison of the mechanical properties of intact cells with those of the purified cytoskeletal biopolymers that are thought to dominate their elasticity reveal the extent to which the studies of purified systems can account for the mechanical properties of the much more heterogeneous and complex cell. This review summarizes selected aspects of current work on cell mechanics with an emphasis on the structures that are activated in cell-cell contacts, that regulate ion flow across the plasma membrane, and that may sense fluid flow that produces low levels of shear stress.
Collapse
Affiliation(s)
- Paul A Janmey
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| | | |
Collapse
|
23
|
Ziober AF, Falls EM, Ziober BL. The extracellular matrix in oral squamous cell carcinoma: friend or foe? Head Neck 2006; 28:740-9. [PMID: 16649214 DOI: 10.1002/hed.20382] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Oral squamous cell carcinoma is a disfiguring, highly invasive and metastatic cancer. Despite advances in detection and therapy, many patients will continue to face a poor prognosis. It is well established that the predominate factor determining overall survival in patients with oral squamous cell carcinoma is lymph node involvement. Tumor growth and progression to invasive cancer requires tumor cell interactions with the extracellular matrix. An understanding of how the extracellular matrix influences tumor development and invasion is fundamental in the development of new prognostic indicators and treatment strategies for oral squamous cell carcinoma. In this review, we summarize how changes in the extracellular matrix contribute to oral cancer development.
Collapse
Affiliation(s)
- Amy F Ziober
- Department of Otorhinolaryngology-Head and Neck Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, 5 Ravdin Building, Philadelphia PA 19104, USA.
| | | | | |
Collapse
|
24
|
Kawano K, Yanagisawa S. Predictive value of laminin-5 and membrane type 1-matrix metalloproteinase expression for cervical lymph node metastasis in T1 and T2 squamous cell carcinomas of the tongue and floor of the mouth. Head Neck 2006; 28:525-33. [PMID: 16619276 DOI: 10.1002/hed.20349] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Laminin-5 (Ln-5) cleaved by membrane type 1-matrix metalloproteinase (MT1-MMP) enhances the migration of tumor cells. The purpose of this study was to determine whether or not enhanced expression of both Ln-5 and MT1-MMP was associated with lymph node metastasis in patients with T1 and T2 squamous cell carcinoma (SCC) of the tongue and floor of the mouth. METHODS By use of biopsy specimens of primary tumors from 57 patients, intratumoral expression of Ln-5 and MT1-MMP was evaluated immunohistochemically and its association with node metastasis analyzed. RESULTS The tumors were categorized into three groups: Ln-5 focal type/MT1-MMP (-) (group I, n = 14), Ln-5 focal type/ MT1-MMP (+) and Ln-5 diffuse type/MT1-MMP (-) (group II, n = 16), and Ln-5 diffuse type/MT1-MMP (+) (group III, n = 27). The incidence of node metastasis (initial and latent metastases) was two of 14 (14.3%), five of 16 (31.3%), and 15 of 27 (55.6%) in groups I, II, and III, respectively. Multivariate analysis identified tumor thickness (odds ratio, 4.751; p = .0152) and Ln-5/ MT1-MMP expression (odds ratio, 3.795, p = .0304) as independent factors of node metastasis. Moreover, in 35 patients with N0 disease, Ln-5/MT1-MMP expression was the only parameter associated with latent node metastasis (odds ratio, 12.800, p = .0247). CONCLUSION These results suggest that immunohistochemical evaluation of Ln-5 and MT1-MMP expression is useful for identifying patients with T1 and T2 SCC of the tongue and floor of the mouth who should be treated with elective neck dissection.
Collapse
Affiliation(s)
- Kenji Kawano
- Department of Oncological Science, Division of Oral and Maxillo-Facial Surgery, Faculty of Medicine, Oita University, 1 Idaigaoka, Hasama-machi, Oita 879-5593, Japan.
| | | |
Collapse
|
25
|
Winterwood NE, Varzavand A, Meland MN, Ashman LK, Stipp CS. A critical role for tetraspanin CD151 in alpha3beta1 and alpha6beta4 integrin-dependent tumor cell functions on laminin-5. Mol Biol Cell 2006; 17:2707-21. [PMID: 16571677 PMCID: PMC1474805 DOI: 10.1091/mbc.e05-11-1042] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2005] [Revised: 03/09/2006] [Accepted: 03/20/2006] [Indexed: 01/07/2023] Open
Abstract
The basement membrane protein laminin-5 supports tumor cell adhesion and motility and is implicated at multiple steps of the metastatic cascade. Tetraspanin CD151 engages in lateral, cell surface complexes with both of the major laminin-5 receptors, integrins alpha3beta1 and alpha6beta4. To determine the role of CD151 in tumor cell responses to laminin-5, we used retroviral RNA interference to efficiently silence CD151 expression in epidermal carcinoma cells. Near total loss of CD151 had no effect on steady state cell surface expression of alpha3beta1, alpha6beta4, or other integrins with which CD151 associates. However, CD151-silenced carcinoma cells displayed markedly impaired motility on laminin-5, accompanied by unusually persistent lateral and trailing edge adhesive contacts. CD151 silencing disrupted alpha3beta1 integrin association with tetraspanin-enriched microdomains, reduced the bulk detergent extractability of alpha3beta1, and impaired alpha3beta1 internalization in cells migrating on laminin-5. Both alpha3beta1- and alpha6beta4-dependent cell adhesion to laminin-5 were also impaired in CD151-silenced cells. Reexpressing CD151 in CD151-silenced cells reversed the adhesion and motility defects. Finally, loss of CD151 also impaired migration but not adhesion on substrates other than laminin-5. These data show that CD151 plays a critical role in tumor cell responses to laminin-5 and reveal promotion of integrin recycling as a novel potential mechanism whereby CD151 regulates tumor cell migration.
Collapse
Affiliation(s)
- Nicole E. Winterwood
- *University of Iowa, Department of Biological Sciences, Iowa City, IA 52240; and
| | - Afshin Varzavand
- *University of Iowa, Department of Biological Sciences, Iowa City, IA 52240; and
| | - Marit N. Meland
- *University of Iowa, Department of Biological Sciences, Iowa City, IA 52240; and
| | - Leonie K. Ashman
- School of Biomedical Sciences, Medical Sciences Building, University of Newcastle, Callaghan NSW 2308, Australia
| | - Christopher S. Stipp
- *University of Iowa, Department of Biological Sciences, Iowa City, IA 52240; and
| |
Collapse
|
26
|
Shintani Y, Wheelock MJ, Johnson KR. Phosphoinositide-3 kinase-Rac1-c-Jun NH2-terminal kinase signaling mediates collagen I-induced cell scattering and up-regulation of N-cadherin expression in mouse mammary epithelial cells. Mol Biol Cell 2006; 17:2963-75. [PMID: 16624865 PMCID: PMC1483033 DOI: 10.1091/mbc.e05-12-1123] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
During epithelial-to-mesenchymal transitions (EMTs), cells must change their interactions with one another and with their extracellular matrix in a synchronized manner. To characterize signaling pathways cells use to coordinate these changes, we used NMuMG mammary epithelial cells. We showed that these cells become fibroblastic and scattered, with increased N-cadherin expression when cultured on collagen I. Rac1 and c-Jun NH2-terminal kinase (JNK) were activated when cells were plated on collagen I, and dominant inhibitory Rac1 (RacN17) or inhibition of JNK signaling prevented collagen I-induced morphological changes and N-cadherin up-regulation. Furthermore, inhibiting phosphoinositide-3 kinase (PI3K) activity prevented Rac1 and JNK activation as well as collagen I-induced N-cadherin up-regulation. These data implicate PI3K-Rac1-JNK signaling in collagen I-induced changes in NMuMG cells. To establish a role for N-cadherin in collagen I-induced cell scattering, we generated N-cadherin overexpressing and knockdown NMuMG cells and showed that knocking down N-cadherin expression prevented collagen I-induced morphological changes. Motility assays showed that cells overexpressing N-cadherin were significantly more motile than mock-transfected cells and that N-cadherin-mediated motility was collagen I dependent. In addition, we showed that cord formation and branching in three-dimensional culture (EMT-dependent events) required N-cadherin expression and PI3K-Rac1-JNK signaling.
Collapse
Affiliation(s)
| | - Margaret J. Wheelock
- Departments of *Oral Biology
- Biochemistry and Molecular Biology
- Genetics, Cell Biology, and Anatomy, and
- Pathology and Microbiology
- Eppley Institute for Research in Cancer and Allied Diseases, and
- Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-7696
| | - Keith R. Johnson
- Departments of *Oral Biology
- Biochemistry and Molecular Biology
- Genetics, Cell Biology, and Anatomy, and
- Pathology and Microbiology
- Eppley Institute for Research in Cancer and Allied Diseases, and
- Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-7696
| |
Collapse
|
27
|
Bourguignon LYW, Gilad E, Brightman A, Diedrich F, Singleton P. Hyaluronan-CD44 interaction with leukemia-associated RhoGEF and epidermal growth factor receptor promotes Rho/Ras co-activation, phospholipase C epsilon-Ca2+ signaling, and cytoskeleton modification in head and neck squamous cell carcinoma cells. J Biol Chem 2006; 281:14026-40. [PMID: 16565089 DOI: 10.1074/jbc.m507734200] [Citation(s) in RCA: 175] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In this study we have examined the interaction of CD44 (a major hyaluronan (HA) receptor) with a RhoA-specific guanine nucleotide exchange factor (leukemia-associated RhoGEF (LARG)) in human head and neck squamous carcinoma cells (HNSCC-HSC-3 cell line). Immunoprecipitation and immunoblot analyses indicate that CD44 and the LARG protein are expressed in HSC-3 cells and that these two proteins are physically associated as a complex. HA-CD44 binding induces LARG-specific RhoA signaling and phospholipase C epsilon (PLC epsilon) activity. In particular, the activation of RhoA-PLC epsilon by HA stimulates inositol 1,4,5-triphosphate production, intracellular Ca2+ mobilization, and the up-regulation of Ca2+/calmodulin-dependent kinase II (CaMKII), leading to phosphorylation of the cytoskeletal protein, filamin. The phosphorylation of filamin reduces its interaction with filamentous actin, promoting tumor cell migration. The CD44-LARG complex also interacts with the EGF receptor (EGFR). Most importantly, the binding of HA to the CD44-LARG-EGFR complex activates the EGFR receptor kinase, which in turn promotes Ras-mediated stimulation of a downstream kinase cascade including the Raf-1 and ERK pathways leading to HNSCC cell growth. Using a recombinant fragment of LARG (the LARG-PDZ domain) and a binding assay, we have determined that the LARG-PDZ domain serves as a direct linker between CD44 and EGFR. Transfection of the HSC-3 cells with LARG-PDZcDNA significantly reduces LARG association with CD44 and EGFR. Overexpression of the LARG-PDZ domain also functions as a dominant-negative mutant (similar to the PLC/Ca2+-calmodulin-dependent kinase II (CaMKII) and EGFR/MAPK inhibitor effects) to block HA/CD44-mediated signaling events (e.g. EGFR kinase activation, Ras/RhoA co-activation, Raf-ERK signaling, PLC epsilon-mediated inositol 1,4,5-triphosphate production, intracellular Ca2+ mobilization, CaMKII activity, filamin phosphorylation, and filamin-actin binding) and to abrogate tumor cell growth/migration. Taken together, our findings suggest that CD44 interaction with LARG and EGFR plays a pivotal role in Rho/Ras co-activation, PLC epsilon-Ca2+ signaling, and Raf/ERK up-regulation required for CaMKII-mediated cytoskeleton function and in head and neck squamous cell carcinoma progression.
Collapse
Affiliation(s)
- Lilly Y W Bourguignon
- Department of Medicine, University of California at San Francisco and Endocrine Unit (111N), Veterans Affairs Medical Center, San Francisco, California 94121, USA.
| | | | | | | | | |
Collapse
|
28
|
Brzóska E, Bello V, Darribère T, Moraczewski J. Integrin α3 subunit participates in myoblast adhesion and fusion in vitro. Differentiation 2006; 74:105-18. [PMID: 16533309 DOI: 10.1111/j.1432-0436.2005.00059.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Satellite cells are myogenic precursor cells, participating in growth, and regeneration of skeletal muscles. The proteins that play a role in myogenesis are integrins. In this report, we show that the integrin alpha3 subunit is expressed in quiescent satellite cells and activated myoblasts. We also find that in myoblasts the integrin alpha3 subunit is localized at cell-cell and cell-extracellular matrix contacts. We notice that increase in protein and mRNA encoding the integrin alpha3 subunit accompanies myoblast differentiation. Using double immunofluorescence and immunoprecipitation experiments, we demonstrate that the integrin alpha3 subunit co-localizes with actin, and binds the integrin beta1 subunit and ADAM12, suggesting that the complex alpha3beta1/ADAM12 is probably involved in myoblast fusion. Importantly, overexpression of the full-length integrin alpha3 subunit increases myoblast fusion whereas an antibody against its extracellular domain inhibits fusion. These data demonstrate that the integrin alpha3 subunit may contribute to satellite cell activation and then myoblast adhesion and fusion.
Collapse
Affiliation(s)
- Edyta Brzóska
- Faculty of Biology, Department of Cytology, Warsaw University, 1 Miecznikowa St., 02-096 Warsaw, Poland.
| | | | | | | |
Collapse
|
29
|
Wang Y, Jin G, Miao H, Li JYS, Usami S, Chien S. Integrins regulate VE-cadherin and catenins: dependence of this regulation on Src, but not on Ras. Proc Natl Acad Sci U S A 2006; 103:1774-9. [PMID: 16446427 PMCID: PMC1413667 DOI: 10.1073/pnas.0510774103] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Adhesions of cells to extracellular matrix and adjacent cells are mediated by integrins and VE-cadherin, respectively. Although these adhesion processes play crucial roles in vascular cell migration and angiogenesis, it remains unclear as to how they are coordinated to regulate cellular functions. We report here that integrin engagement by treating bovine endothelial aortic cell monolayers with beads coated with fibronectin (Fn) led to disruption of the VE-cadherin-containing adherens junctions. This disruption was accompanied by increases of tyrosine phosphorylation of beta-catenin, gamma-catenin, and p120ctn, as well as the dissociation of alpha-catenin and gamma-catenin from VE-cadherin. We applied a membrane-targeted Src reporter based on the fluorescence resonance energy transfer technique to visualize the dynamic Src activation at subcellular levels in live cells. The integrin engagement induced by Fn-coated beads caused the activation of Src around the beads and at adherens junctions, which are subsequently disrupted. The inhibition of Src with PP1 blocked the effects of integrin engagement on adherens junctions. Although Ras can also modulate adherens junctions, the resulting patterns of phosphorylation and association of junction proteins were distinct from those induced by integrin engagement. The inhibition of Ras by RasN17 did not rescue the disruption of adherens junctions induced by integrin engagement or by Src activation. Integrin engagement by Fn-coated beads also induced a significant alteration of cortical actin filaments at adherens junctions. The results indicate that integrin engagement disrupts VE-cadherin-containing adherens junctions via the activation of Src, but not Ras, possibly as a result of modulation of the actin network.
Collapse
Affiliation(s)
- Yingxiao Wang
- Departments of Bioengineering and Medicine and The Whitaker Institute of Biomedical Engineering, University of California at San Diego, La Jolla, CA 92093
| | - Gang Jin
- Departments of Bioengineering and Medicine and The Whitaker Institute of Biomedical Engineering, University of California at San Diego, La Jolla, CA 92093
| | - Hui Miao
- Departments of Bioengineering and Medicine and The Whitaker Institute of Biomedical Engineering, University of California at San Diego, La Jolla, CA 92093
| | - Julie Y.-S. Li
- Departments of Bioengineering and Medicine and The Whitaker Institute of Biomedical Engineering, University of California at San Diego, La Jolla, CA 92093
| | - Shunichi Usami
- Departments of Bioengineering and Medicine and The Whitaker Institute of Biomedical Engineering, University of California at San Diego, La Jolla, CA 92093
| | - Shu Chien
- Departments of Bioengineering and Medicine and The Whitaker Institute of Biomedical Engineering, University of California at San Diego, La Jolla, CA 92093
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
30
|
Burleson KM, Boente MP, Pambuccian SE, Skubitz APN. Disaggregation and invasion of ovarian carcinoma ascites spheroids. J Transl Med 2006; 4:6. [PMID: 16433903 PMCID: PMC1397876 DOI: 10.1186/1479-5876-4-6] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2005] [Accepted: 01/24/2006] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Malignant ascites often develops in advanced stages of ovarian carcinoma, consisting of single and aggregated tumor cells, or spheroids. Spheroids have commonly been used as tumor models to study drug efficacy, and have shown resistance to some chemotherapies and radiation. However, little is known about the adhesive or invasive capabilities of spheroids, and whether this particular cellular component of the ascites can contribute to dissemination of ovarian cancer. Here, we examined the invasive ability of ascites spheroids recovered from seven ovarian carcinoma patients and one primary peritoneal carcinoma (PPC) patient. METHODS Ascites spheroids were isolated from patients, purified, and immunohistochemical analyses were performed by a pathologist to confirm diagnosis. In vitro assays were designed to quantify spheroid disaggregation on a variety of extracellular matrices and dissemination on and invasion into normal human mesothelial cell monolayers. Cell proliferation and viability were determined in each assay, and statistical significance demonstrated by the student's t-test. RESULTS Spheroids from all of the patients' ascites samples disaggregated on extracellular matrix components, with the PPC spheroids capable of complete disaggregation on type I collagen. Additionally, all of the ascites spheroid samples adhered to and disaggregated on live human mesothelial cell monolayers, typically without invading them. However, the PPC ascites spheroids and one ovarian carcinoma ascites spheroid sample occasionally formed invasive foci in the mesothelial cell monolayers, suggestive of a more invasive phenotype. CONCLUSION We present here in vitro assays using ascites spheroids that imitate the spread of ovarian cancer in vivo. Our results suggest that systematic studies of the ascites cellular content are necessary to understand the biology of ovarian carcinoma.
Collapse
Affiliation(s)
- Kathryn M Burleson
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Matthew P Boente
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stefan E Pambuccian
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Amy PN Skubitz
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
31
|
Burleson KM, Hansen LK, Skubitz APN. Ovarian carcinoma spheroids disaggregate on type I collagen and invade live human mesothelial cell monolayers. Clin Exp Metastasis 2005; 21:685-97. [PMID: 16035613 DOI: 10.1007/s10585-004-5768-5] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Ovarian carcinoma patients frequently develop malignant ascites containing single and aggregated tumor cells, or spheroids. Spheroids have been shown to be resistant to many therapies, but their contribution to ovarian cancer dissemination remains undetermined. We have previously shown that ascites spheroids adhere to extracellular matrix (ECM) proteins and live human mesothelial cells via beta1 integrin subunits. Here, we assessed the ability of spheroids that were generated from the human ovarian carcinoma cell line NIH:OVCAR5 to disseminate and invade in vitro. Spheroids were seeded on ECM proteins for 24 h. While laminin and type IV collagen stimulated some cell migration, spheroids completely disaggregated on type I collagen substrates. A monoclonal antibody against the beta1 integrin subunit significantly inhibited disaggregation on all proteins tested. To test their invasive ability, spheroids were added to monolayers of live human LP9 mesothelial cells. Within 24 h, the spheroids adhered and disaggregated on top of the monolayers, and within a week had established foci of invasion encompassing a 200-fold larger surface area. Addition of a monoclonal antibody against the beta1 integrin subunit drastically reduced spheroid invasion into the mesothelial cell monolayers. GM 6001, a broad-scale matrix metalloproteinase inhibitor, also significantly blocked spheroid invasion into the mesothelial cell monolayers. Epsilon-amino-N-caproic acid, a serine protease inhibitor, partially inhibited spheroid invasion. Based on their ability to attach to, disaggregate on, and invade into live human mesothelial cell monolayers, spheroids should thus be regarded as potential contributors to the dissemination of ovarian cancer.
Collapse
Affiliation(s)
- Kathryn M Burleson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
32
|
Abstract
Squamous cell carcinoma (SCC) is the primary tumor type in head and neck cancer. Typically, these tumor cells show persistent invasion that frequently leads to local recurrence and distant lymphatic metastasis. The process of invasion involves concurrent infiltration and destruction of adjacent tissues. As with normal mucosal epithelium, SCC cells express receptors that mediate cell-extracellular matrix (ECM) adhesion (integrins) and cell-cell adhesion (cadherins). Both receptor families represent important signaling devices that are capable of promoting survival and proliferation. Recent results indicate that integrins and cadherins cooperate to regulate invasive behavior. During SCC invasion, cells actively migrate through the surrounding ECM with the simultaneous remodeling of their intercellular adhesions. During invasion, integrin receptor engagement with specific ECM ligands along with concurrent remodeling of cadherin adhesions induces changes in the cytoskeleton though modulation of the activities of Rho family members. Tumor development and progression of SCC proceeds with the generation of variant cells with potential alterations in expression of adhesion receptors, and their associated signaling pathways lead to a highly invasive and metastatic phenotype. Understanding the molecular events that define this subset of invasive cells will facilitate the development of new treatment strategies.
Collapse
Affiliation(s)
- Randall H Kramer
- Head and Neck Oncology Program, Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA.
| | | | | |
Collapse
|
33
|
Zhou H, Kramer RH. Integrin engagement differentially modulates epithelial cell motility by RhoA/ROCK and PAK1. J Biol Chem 2005; 280:10624-35. [PMID: 15611088 DOI: 10.1074/jbc.m411900200] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Integrin-ligand binding regulates tumor cell motility and invasion. Cell migration also involves the Rho GTPases that control the interplay between adhesion receptors and the cytoskeleton. We evaluated how specific extracellular matrix ligands modulate Rho GTPases and control motility of human squamous cell carcinoma cells. On laminin-5 substrates, the epithelial cells rapidly spread and migrated, but on type I collagen the cells spread slowly and showed reduced motility. We found that RhoA activity was suppressed in cells attached to laminin-5 through the alpha3 integrin receptor. In contrast, RhoA was strongly activated in cells bound to type I collagen and this was mediated by the alpha2 integrin. Inhibiting the RhoA pathway by expression of a dominant-negative RhoA mutant or by directly inhibiting ROCK, reduced focal adhesion formation and enhanced cell migration on type I collagen. Cdc42 and Rac and their downstream target PAK1 were activated following adhesion to laminin-5. PAK1 activation induced by laminin-5 was suppressed by expression of a dominant-negative Cdc42. Moreover, constitutively active PAK1 stimulated migration on collagen I substrates. Our results indicate that in squamous epithelial cells, collagen-alpha2beta1 integrin binding activates RhoA, slowing cell locomotion, whereas laminin-5-alpha3beta1 integrin interaction inhibits RhoA and activates PAK1, stimulating cell migration. The data demonstrate that specific ligand-integrin pairs regulate cell motility differentially by selectively modulating activities of Rho GTPases and their effectors.
Collapse
Affiliation(s)
- Hua Zhou
- Department of Cell and Tissue Biology, University of California, San Francisco, California 94143, USA
| | | |
Collapse
|
34
|
Hintermann E, Yang N, O'Sullivan D, Higgins JMG, Quaranta V. Integrin alpha6beta4-erbB2 complex inhibits haptotaxis by up-regulating E-cadherin cell-cell junctions in keratinocytes. J Biol Chem 2005; 280:8004-15. [PMID: 15579904 DOI: 10.1074/jbc.m406301200] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Keratinocyte integrins alpha6beta4 and alpha3beta1 bind laminin-5, a component of basement membranes. We previously demonstrated that in keratinocytes, haptotactic migration on laminin-5 was stimulated by anti-beta1 integrin-activating antibody TS2/16, whereas antibodies to alpha6 and beta4, respectively, blocked TS2/16-induced, alpha3beta1-dependent migration. Moreover, alpha6beta4-associated haptotaxis inhibition was linked to a phosphatidylinositol 3-kinase (PI3K) pathway and required erbB2 activation. erbB2, the ligand-less member of the epidermal growth factor receptor family, was shown to form a complex with the hemidesmosomal integrin alpha6beta4. Here, we demonstrate that alpha6beta4 inhibitory effects on haptotaxis are abolished by an anti-E-cadherin antibody, which interferes with cell-cell adhesion. Furthermore, antibodies to alpha6 and beta4 stimulated adhesion to an E-cadherin-Fc recombinant protein. In addition, anti-alpha6/beta4 antibodies increased colony size in plated cells, stimulated cell-cell aggregation, and up-regulated E-cadherin localization to cell-cell contacts. These effects were abolished when erbB2 or PI3K were blocked. These results indicate that stimulation of alpha6beta4 increases E-cadherin-mediated cell-cell adhesion and that this mechanism depends on erbB2 activation. The molecule that links alpha6beta4 with E-cadherin may be the small GTPase cdc42, an effector of PI3K, because dominant-negative cdc42 abolished the inhibitory effect of anti-alpha6/beta4 antibodies and increased basal migration, whereas constitutively active cdc42 prevented the TS2/16-induced increase in haptotaxis. These findings suggest a model whereby alpha6beta4 can augment cell-cell adhesion and slow down haptotaxis over laminin-5 and point to the alpha6beta4-erbB2 heterodimer as an important signaling complex for the formation of cohesive keratinocyte layers.
Collapse
Affiliation(s)
- Edith Hintermann
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
35
|
Shen X, Kramer RH. Adhesion-mediated squamous cell carcinoma survival through ligand-independent activation of epidermal growth factor receptor. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:1315-29. [PMID: 15466396 PMCID: PMC1618631 DOI: 10.1016/s0002-9440(10)63390-1] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The survival and growth of squamous epithelial cells require signals generated by integrin-matrix interactions. After conversion to squamous cell carcinoma, the cells remain sensitive to detachment-induced anoikis, yet in tumor cell aggregates, which are matrix-deficient, these cells are capable of suprabasal survival and proliferation. Their survival is enhanced through a process we call synoikis, whereby junctional adhesions between neighboring cells generate specific downstream survival signals. Here we show that in squamous cell carcinoma cells, E-cadherin-mediated cell-cell contacts specifically induce activation of epidermal growth factor receptor (EGFR). EGFR activation in turn triggers the ERK/MAPK signaling module, leading to elevation of anti-apoptotic Bcl-2. After intercellular adhesion, formation of adherens junctions triggers the formation of E-cadherin-EGFR complexes, correlating with EGFR transactivation. Analysis of the process with a dominant-negative EGFR mutant indicated that activation of EGFR is ligand-independent. Our data implicate cell-cell adhesion-induced activation of EGFR as a cooperative mechanism that generates compensatory survival signaling, protecting malignant cells from detachment-induced death.
Collapse
Affiliation(s)
- Xiaodong Shen
- Department of Stomatology, School of Medicine, University of California at San Francisco, Box 0512, Room HSW-604, San Francisco, CA 94143-0512, USA
| | | |
Collapse
|
36
|
Burleson KM, Casey RC, Skubitz KM, Pambuccian SE, Oegema TR, Skubitz APN. Ovarian carcinoma ascites spheroids adhere to extracellular matrix components and mesothelial cell monolayers. Gynecol Oncol 2004; 93:170-81. [PMID: 15047232 DOI: 10.1016/j.ygyno.2003.12.034] [Citation(s) in RCA: 200] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2003] [Indexed: 01/02/2023]
Abstract
OBJECTIVES Ovarian carcinoma cells form multicellular aggregates, or spheroids, in the peritoneal cavity of patients with advanced disease. The current paradigm that ascites spheroids are non-adhesive leaves their contribution to ovarian carcinoma dissemination undefined. Here, spheroids obtained from ovarian carcinoma patients' ascites were characterized for their ability to adhere to molecules encountered in the peritoneal cavity, with the goal of establishing their potential to contribute to ovarian cancer spread. METHODS Spheroids were recovered from the ascites fluid of 11 patients with stage III or stage IV ovarian carcinoma. Adhesion assays to extracellular matrix (ECM) proteins and human mesothelial cell monolayers were performed for each of the ascites spheroid samples. Subsequently, inhibition assays were performed to identify the cell receptors involved. RESULTS Most ascites samples adhered moderately to fibronectin and type I collagen, with reduced adhesion to type IV collagen and laminin. Monoclonal antibodies against the beta1 integrin subunit partially inhibited this adhesion. Ascites spheroids also adhered to hyaluronan. Additionally, spheroids adhered to live, but not fixed, human mesothelial cell monolayers, and this adhesion was partially mediated by beta1 integrins. CONCLUSIONS The cellular content of the ascites fluid has often been considered non-adhesive, but our findings are the first to suggest that patient-derived ascites spheroids can adhere to mesothelial extracellular matrix via beta1 integrins, indicating that spheroids should not be ignored in the dissemination of ovarian cancer.
Collapse
Affiliation(s)
- Kathryn M Burleson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | |
Collapse
|
37
|
Stipp CS, Kolesnikova TV, Hemler ME. EWI-2 regulates alpha3beta1 integrin-dependent cell functions on laminin-5. J Cell Biol 2003; 163:1167-77. [PMID: 14662754 PMCID: PMC2173626 DOI: 10.1083/jcb.200309113] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2003] [Accepted: 10/14/2003] [Indexed: 12/16/2022] Open
Abstract
EWI-2, a cell surface immunoglobulin SF protein of unknown function, associates with tetraspanins CD9 and CD81 with high stoichiometry. Overexpression of EWI-2 in A431 epidermoid carcinoma cells did not alter cell adhesion or spreading on laminin-5, and had no effect on reaggregation of cells plated on collagen I (alpha2beta1 integrin ligand). However, on laminin-5 (alpha3beta1 integrin ligand), A431 cell reaggregation and motility functions were markedly impaired. Immunodepletion and reexpression experiments revealed that tetraspanins CD9 and CD81 physically link EWI-2 to alpha3beta1 integrin, but not to other integrins. CD81 also controlled EWI-2 maturation and cell surface localization. EWI-2 overexpression not only suppressed cell migration, but also redirected CD81 to cell filopodia and enhanced alpha3beta1-CD81 complex formation. In contrast, an EWI-2 chimeric mutant failed to suppress cell migration, redirect CD81 to filopodia, or enhance alpha3beta1-CD81 complex formation. These results show how laterally associated EWI-2 might regulate alpha3beta1 function in disease and development, and demonstrate how tetraspanin proteins can assemble multiple nontetraspanin proteins into functional complexes.
Collapse
Affiliation(s)
- Christopher S Stipp
- Dana-Farber Cancer Institute, Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|
38
|
Taddei I, Faraldo MM, Teulière J, Deugnier MA, Thiery JP, Glukhova MA. Integrins in mammary gland development and differentiation of mammary epithelium. J Mammary Gland Biol Neoplasia 2003; 8:383-94. [PMID: 14985635 DOI: 10.1023/b:jomg.0000017426.74915.b9] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Integrins are major extracellular matrix (ECM) receptors that can also serve for some cell-cell interactions. They have been identified as important regulators of mammary epithelial cell growth and differentiation. Their ability to promote cell anchorage, proliferation, survival, migration, and the induction of active ECM-degrading enzymes suggests that they play an essential role in normal mammary morphogenesis, but, on the other hand, reveals their potential to promote tumor progression.
Collapse
Affiliation(s)
- Ilaria Taddei
- UMR 144 CNRS-Institut Curie, Section de Recherche, 26, rue d'Ulm, 75248 Paris Cedex 05, France
| | | | | | | | | | | |
Collapse
|
39
|
Martinez-Lemus LA, Wu X, Wilson E, Hill MA, Davis GE, Davis MJ, Meininger GA. Integrins as unique receptors for vascular control. J Vasc Res 2003; 40:211-33. [PMID: 12902635 DOI: 10.1159/000071886] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2002] [Accepted: 04/01/2003] [Indexed: 11/19/2022] Open
Abstract
Cells within the vascular wall connect their cytoskeleton to the extracellular matrix (ECM) through a family of cell surface receptors known as integrins. The ability of integrins to act as a link between the extracellular and intracellular environments allows transmission of inside-out and outside-in signals capable of modulating diverse vascular phenomena. In this review we summarize what is currently known about the involvement of integrins in the control of vascular tone, permeability and remodeling. We discuss the capacity of integrins to act as detectors of injury-generated molecules derived from ECM proteins, as well as the putative role of integrins as mechanosensors for shear and tension. Particular attention is given to the mechanisms responsible for linking integrins to the control of vascular tone, and we review the intracellular signaling pathways involved in effecting the vascular responses elicited by integrin activation. Finally, the involvement of integrins in vascular remodeling and vascular disease is analyzed. Considerable evidence strongly indicates that integrins are involved in both acute and chronic vascular control. Understanding the elements and the sequence of events linking integrins with vasoregulation is important for deciphering phenomena such as the pressure-dependent myogenic response, flow-dependent changes in vascular diameter, and vascular remodeling as they occur in physiological and pathological conditions. Further understanding of the role of integrins in vascular control holds promise as new avenues for prophylactic and therapeutic manipulation of vascular phenomena.
Collapse
Affiliation(s)
- Luis A Martinez-Lemus
- Department of Medical Physiology, Cardiovascular Research Institute, Texas A&M University System Health Science Center, College Station, Texas 77843-1114, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Levy Y, Ronen D, Bershadsky AD, Zick Y. Sustained induction of ERK, protein kinase B, and p70 S6 kinase regulates cell spreading and formation of F-actin microspikes upon ligation of integrins by galectin-8, a mammalian lectin. J Biol Chem 2003; 278:14533-42. [PMID: 12569102 DOI: 10.1074/jbc.m207380200] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Galectin-8, a member of the galectin family of mammalian lectins, is a secreted protein that promotes cell adhesion and migration upon binding to a subset of integrins through sugar-protein interactions. Ligation of integrins by galectin-8 triggers a distinct pattern of cytoskeletal organization, including formation of F-actin-containing microspikes. This is associated with activation of integrin-mediated signaling cascades (ERK and phosphatidylinositol 3 kinase (PI3K)) that are much more robust and are of longer duration than those induced upon cell adhesion to fibronectin. Indeed, formation of microspikes is enhanced 40% in cells that overexpress protein kinase B, the downstream effector of PI3K. Inhibition of PI3K activity induced by wortmannin partially inhibits cell adhesion and spreading while largely inhibiting microspike formation in cells adherent to galectin-8. Furthermore, the inhibitory effects of wortmannin are markedly accentuated in cells overexpressing PKB or p70S6K (CHO(PKB) and CHO(p70S6K) cells), whose adhesion and spreading on galectin-8 (but not on fibronectin) is inhibited approximately 25-35% in the presence of wortmannin. The above results suggest that galectin-8 is an extracellular matrix protein that triggers a unique repertoire of integrin-mediated signals, which leads to a distinctive cytoskeletal organization and microspike formation. They further suggest that downstream effectors of PI3K, including PKB and p70 S6 kinase, in part mediate cell adhesion, spreading, and microspike formation induced by galectin-8.
Collapse
Affiliation(s)
- Yifat Levy
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | |
Collapse
|
41
|
Pelosi G, Pastorino U, Pasini F, Maissoneuve P, Fraggetta F, Iannucci A, Sonzogni A, De Manzoni G, Terzi A, Durante E, Bresaola E, Pezzella F, Viale G. Independent prognostic value of fascin immunoreactivity in stage I nonsmall cell lung cancer. Br J Cancer 2003; 88:537-47. [PMID: 12592367 PMCID: PMC2377175 DOI: 10.1038/sj.bjc.6600731] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Fascin-1, the most expressed form of fascin in vertebrate tissues, is an actin-bundling protein that induces cell membrane protrusions and increases motility of normal and transformed epithelial cells. Very few data are available on the role of this protein in nonsmall cell lung cancer (NSCLC). Two hundred and twenty patients with stage I NSCLC and long-term follow-up were evaluated immunocytochemically for fascin expression. Overall, variable fascin immunoreactivity was detected in 98% of 116 squamous cell carcinomas, in 78% of 96 adenocarcinomas, in 83% of six large cell carcinomas, and in the two adenosquamous carcinomas under study. Neoplastic emboli were commonly decorated by the antifascin antibody (P<0.001), also when the surrounding invasive carcinoma was unreactive. Fascin immunoreactivity correlated with high tumour grade (P=0.017) and, in adenocarcinomas, with high Ki-67 labelling index (P=0.021). Adenocarcinomas with a prevalent bronchiolo-alveolar in situ component were less commonly immunoreactive for fascin than invasive tumours (P=0.005). Contralateral thoracic or distant metastases were associated significantly with diffuse (>60% immunoreactive tumour cells) fascin expression in adenocarcinomas (P=0.043), and marginally with strong fascin immunostaining in squamous cell carcinomas (P=0.13). No associations were noted with any other clinicopathological variables tested. Patients with tumours showing diffuse (>60% immunoreactive neoplastic cells) and/or strong immunoreactivity for fascin had a shorter survival (P=0.006 for adenocarcinomas and P=0.026 for squamous cell carcinomas), even after multivariate analysis (P=0.014 and 0.050, respectively). The current study documents for the first time that fascin is upregulated in invasive and more aggressive NSCLC, being an independent prognostic predictor of unfavourable clinical course of the disease. Targetting the fascin pathway could be a novel therapeutic strategy of NSCLC.
Collapse
Affiliation(s)
- G Pelosi
- Department of Pathology and Laboratory Medicine, University of Milan School of Medicine, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Pakkala T, Virtanen I, Oksanen J, Jones JCR, Hormia M. Function of laminins and laminin-binding integrins in gingival epithelial cell adhesion. J Periodontol 2002; 73:709-19. [PMID: 12146529 DOI: 10.1902/jop.2002.73.7.709] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND In human gingiva, epithelial cells attach to their adjacent tissues by means of specialized molecular adhesion complexes and a basement membrane. Little is known about the synthesis of adhesion proteins by gingival keratinocytes; we, therefore, studied how cultured immortalized gingival epithelial cells produce laminins and express laminin-binding integrins. We presumed that different laminins and integrins would be involved in the adhesion of gingival epithelial cells. METHODS We cultured gingival keratinocytes and studied their production of laminins and expression of integrins using immunofluorescence microscopy, immunoprecipitation, and immunoblotting methods and by quantitative cell adhesion experiments. We also studied how gingival tissue expresses these adhesion proteins in vivo by using immunofluorescence microscopy. RESULTS In immunofluorescence microscopy, the cells were seen to organize chains of laminin-5 (alpha3beta03gamma2) to extracellular patches, whereas the alpha5 chain of laminin-10 (alpha5betalgamma1) could only be seen intracellularly. Of the laminin-binding integrin subunits, integrin a6 subunit was organized to dotted arrays, typical of prehemidesmosomal adhesions, whereas integrin alpha3 subunit was located at cell-cell junctions, in prehemidesmosomal structures, and at some locations also in small focal-contact like patches. Integrin beta1 subunit was found at cell-cell junctions and in focal contacts. Immunoprecipitation experiments showed that the cells synthesize and secrete chains of laminin-5 and laminin-10. In quantitative cell adhesion experiments, the cells adhered efficiently to these laminins by using cooperatively integrin alpha3beta1 and alpha6beta1 integrin complexes. None of the other known laminin-binding integrin subunits appeared to be significantly involved in cell adhesion to these laminin isoforms. CONCLUSIONS Our results provide new information on gingival epithelial cell adhesion and extracellular matrix production and may thus aid in the understanding of periodontal physiology.
Collapse
Affiliation(s)
- Tuomas Pakkala
- Institute of Biomedicine/Anatomy, Biomedicum Helsinki, University of Helsinki, Finland.
| | | | | | | | | |
Collapse
|
43
|
Patarroyo M, Tryggvason K, Virtanen I. Laminin isoforms in tumor invasion, angiogenesis and metastasis. Semin Cancer Biol 2002; 12:197-207. [PMID: 12083850 DOI: 10.1016/s1044-579x(02)00023-8] [Citation(s) in RCA: 271] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Laminins are a growing family of alphabetagamma heterotrimeric proteins, commonly found in basement membranes (BMs). These large molecules promote cell adhesion and migration via integrins and other cell-surface receptors. Over 12 laminin isoforms are presently known. The various isoforms have a cell- and tissue-specific expression and are differentially recognized by integrins. Expression of laminin isoforms in tumors usually reflects expression in their normal counterparts. However, during tumor invasion, loss of the BM barrier occurs and a discontinuous pattern of laminin staining is observed. In carcinomas, tumor cells at the invading front strongly express intracellularly the gamma2 chain, a component of laminin-5. Remodeling of the vascular BM is observed during angiogenesis, and penetration of several BMs occurs during tumor dissemination and metastasis. Thus, disregulated cell-laminin interactions are major traits of malignant disorders.
Collapse
Affiliation(s)
- Manuel Patarroyo
- Microbiology and Tumorbiology Center/Odontology, Karolinska Institutet, S 171 77 Stockholm, Sweden.
| | | | | |
Collapse
|
44
|
Affiliation(s)
- Alex Lopata
- Department of Obstetrics and Gynecology, University of Melbourne, Royal Women's Hospital, Carlton, Victoria 3053, Australia.
| | | | | |
Collapse
|
45
|
Kureishy N, Sapountzi V, Prag S, Anilkumar N, Adams JC. Fascins, and their roles in cell structure and function. Bioessays 2002; 24:350-61. [PMID: 11948621 DOI: 10.1002/bies.10070] [Citation(s) in RCA: 260] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The fascins are a structurally unique and evolutionarily conserved group of actin cross-linking proteins. Fascins function in the organisation of two major forms of actin-based structures: dynamic, cortical cell protrusions and cytoplasmic microfilament bundles. The cortical structures, which include filopodia, spikes, lamellipodial ribs, oocyte microvilli and the dendrites of dendritic cells, have roles in cell-matrix adhesion, cell interactions and cell migration, whereas the cytoplasmic actin bundles appear to participate in cell architecture. We discuss the current understanding of the cellular mechanisms that regulate the binding of fascin to actin and how these processes contribute to the organisation or disassembly of cell protrusions. Although the in vivo roles of fascin have been studied principally in Drosophila, several human diseases are associated with inherited or acquired alterations in the expression of fascins. Strategies to modulate fascin-containing protrusions and thereby cell adhesive and migratory behaviour could have potential for therapeutic intervention in these conditions. The supplementary material referred to in this section can be found at http://www.interscience.wiley.com/jpages/0265-9247/suppmat/2002/v24.350.html
Collapse
Affiliation(s)
- Nina Kureishy
- MRC Laboratory for Molecular Cell Biology and Department of Biochemistry and Molecular Biology, University College London
| | | | | | | | | |
Collapse
|
46
|
Chamoux E, Narcy A, Lehoux JG, Gallo-Payet N. Fibronectin, laminin, and collagen IV as modulators of cell behavior during adrenal gland development in the human fetus. J Clin Endocrinol Metab 2002; 87:1819-28. [PMID: 11932324 DOI: 10.1210/jcem.87.4.8359] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The specific development of the human fetal adrenal gland requires cell proliferation, migration, apoptosis, and zone-specific steroidogenic activity. The present work was designed to determine the physiological significance of the previously identified spatial distribution of extracellular matrix components in the fetal gland. Primary cultures of human fetal adrenal cells grown on collagen IV, laminin, or fibronectin revealed that cell morphology was affected by environmental cues. Matrices also modulated the profile of steroid secretion by the fetal cells. Collagen IV favored cortisol secretion after ACTH or angiotensin II stimulation and increased dehydroepiandrosterone production when the AT(2) receptor of angiotensin II was specifically stimulated. These effects were correlated by changes in the mRNA levels of 3beta-hydroxysteroid dehydrogenase and cytochrome P450C17. In contrast, fibronectin and laminin decreased cell responsiveness to ACTH in terms of cortisol secretion, but enhanced ACTH-stimulated androgen secretion. Finally, extracellular matrices were able to orchestrate cell behavior. Collagen IV and laminin enhanced cell proliferation, and fibronectin increased cell death. This study is the first to demonstrate that the nature of extracellular matrix coordinates specific steroidogenic pathways and cell turnover in the developing human fetal adrenal gland.
Collapse
Affiliation(s)
- Estelle Chamoux
- Service of Endocrinology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | | | | | | |
Collapse
|
47
|
Rust WL, Carper SW, Plopper GE. The Promise of Integrins as Effective Targets for Anticancer Agents. J Biomed Biotechnol 2002; 2:124-130. [PMID: 12488576 PMCID: PMC161362 DOI: 10.1155/s1110724302204015] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
This review will briefly describe integrin function, address why integrins are attractive targets for chemotherapeutic drug design, and discuss some ongoing studies aimed at inhibiting integrin activity. Integrins are cell surface heterodimeric receptors. They modulate many cellular processes including: growth, death (apoptosis), adhesion, migration, and invasion by activating several signaling pathways. Many potential chemotherapeutic agents target integrins directly (eg, polypeptides, monoclonal antibodies, adenovirus vectors). These agents may be clinically useful in controlling the metastatic spread of cancer.
Collapse
Affiliation(s)
- William L. Rust
- UNLV Cancer Institute and Chemistry Department, University of Nevada, Las Vegas, NV 89154, USA
| | - Stephen W. Carper
- UNLV Cancer Institute and Chemistry Department, University of Nevada, Las Vegas, NV 89154, USA
| | - George E. Plopper
- Department of Biology, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180-3590, USA
| |
Collapse
|
48
|
Casey RC, Burleson KM, Skubitz KM, Pambuccian SE, Oegema TR, Ruff LE, Skubitz AP. Beta 1-integrins regulate the formation and adhesion of ovarian carcinoma multicellular spheroids. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 159:2071-80. [PMID: 11733357 PMCID: PMC1850600 DOI: 10.1016/s0002-9440(10)63058-1] [Citation(s) in RCA: 188] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Ovarian carcinoma multicellular spheroids are an in vitro model of micrometastasis whose adhesive abilities have not been elucidated. In this study, we identified adhesion molecules that mediate the formation of ovarian carcinoma spheroids and their subsequent adhesion to extracellular matrix proteins. The NIH:OVCAR5, but not the SKOV3, ovarian carcinoma cell line formed spheroids similar to multicellular aggregates isolated from patient ascitic fluid. NIH:OVCAR5 spheroid formation was augmented by a beta 1-integrin-stimulating monoclonal antibody or exogenous fibronectin, but was inhibited by blocking monoclonal antibodies against the alpha 5- or beta 1-integrin subunits. By immunohistochemical staining, alpha 2-, alpha 3-, alpha 5-, alpha 6-, and beta 1-integrin subunits, CD44, and fibronectin were detected in NIH:OVCAR5 spheroids. NIH:OVCAR5 spheroids adhered to fibronectin, laminin, and type IV collagen, and this adhesion was partially inhibited by blocking antibodies against the alpha 5-, alpha 6-, and alpha 2-integrin subunits, respectively. A blocking monoclonal antibody against the beta 1-integrin subunit completely inhibited adhesion of the spheroids to all three proteins. These results suggest that interactions between the alpha 5 beta 1-integrin and fibronectin mediate the formation of ovarian carcinoma spheroids and that their adhesion to extracellular matrix proteins at sites of secondary tumor growth may be mediated by a complex interaction between multiple integrins and their ligands.
Collapse
Affiliation(s)
- R C Casey
- Departments of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Chapin RE, Wine RN, Harris MW, Borchers CH, Haseman JK. Structure and control of a cell-cell adhesion complex associated with spermiation in rat seminiferous epithelium. JOURNAL OF ANDROLOGY 2001; 22:1030-52. [PMID: 11700851 DOI: 10.1002/j.1939-4640.2001.tb03444.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Spermiation, the release of late spermatids from the Sertoli cell, is disrupted by a number of toxicants. Control of the spermiation process, and the proteins that interact to adhere mature spermatids to Sertoli cells, is poorly understood. In these studies we used immunohistochemistry, coimmunoprecipitation/Western blotting, and mass spectrometry to refine an earlier model of sperm adhesion proposed by our laboratory. We have identified specific proteins linked together as part of a multiprotein complex, as well as several additional proteins (cortactin, ERK1/2, and 14-3-3 zeta) that may be functioning in both structural and signal transduction roles. The current and prior data suggest that protein phosphorylation is central to the control of spermiation. We also present and characterize an in vitro tubule culture system that allowed functional testing of the spermiation model by pharmacologic manipulation, and yielded data consistent with the importance of protein phosphorylation in spermiation.
Collapse
Affiliation(s)
- R E Chapin
- Reproductive Toxicology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA.
| | | | | | | | | |
Collapse
|
50
|
Seewaldt VL, Mrózek K, Sigle R, Dietze EC, Heine K, Hockenbery DM, Hobbs KB, Caldwell LE. Suppression of p53 function in normal human mammary epithelial cells increases sensitivity to extracellular matrix-induced apoptosis. J Cell Biol 2001; 155:471-86. [PMID: 11673474 PMCID: PMC2150841 DOI: 10.1083/jcb.200011001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Little is known about the fate of normal human mammary epithelial cells (HMECs) that lose p53 function in the context of extracellular matrix (ECM)-derived growth and polarity signals. Retrovirally mediated expression of human papillomavirus type 16 (HPV-16) E6 and antisense oligodeoxynucleotides (ODNs) were used to suppress p53 function in HMECs as a model of early breast cancer. p53+ HMEC vector controls grew exponentially in reconstituted ECM (rECM) until day 6 and then underwent growth arrest on day 7. Ultrastructural examination of day 7 vector controls revealed acinus-like structures characteristic of normal mammary epithelium. In contrast, early passage p53- HMEC cells proliferated in rECM until day 6 but then underwent apoptosis on day 7. p53- HMEC-E6 passaged in non-rECM culture rapidly (8-10 passages), lost sensitivity to both rECM-induced growth arrest and polarity, and also developed resistance to rECM-induced apoptosis. Resistance was associated with altered expression of alpha3-integrin. Treatment of early passage p53- HMEC-E6 cells with either alpha3- or beta1-integrin function-blocking antibodies inhibited rECM-mediated growth arrest and induction of apoptosis. Our results indicate that suppression of p53 expression in HMECs by HPV-16 E6 and ODNs may sensitize cells to rECM-induced apoptosis and suggest a role for the alpha3/beta1-heterodimer in mediating apoptosis in HMECs grown in contact with rECM.
Collapse
Affiliation(s)
- V L Seewaldt
- Division of Medical Oncology, Duke University, Durham, NC 27710, USA.
| | | | | | | | | | | | | | | |
Collapse
|