1
|
Ranganathan S, Reddy A, Russo A, Malepelle U, Desai A. Double agents in immunotherapy: Unmasking the role of antibody drug conjugates in immune checkpoint targeting. Crit Rev Oncol Hematol 2024; 202:104472. [PMID: 39111458 DOI: 10.1016/j.critrevonc.2024.104472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024] Open
Abstract
Antibody-drug conjugates (ADCs) have high specificity with lesser off-target effects, thus providing improved efficacy over traditional chemotherapies. A total of 14 ADCs have been approved for use against cancer by the US Food and Drug Administration (FDA), with more than 100 ADCs currently in clinical trials. Of particular interest ADCs targeting immune antigens PD-L1, B7-H3, B7-H4 and integrins. Specifically, we describe ADCs in development along with the gene and protein expression of these immune checkpoints across a wide range of cancer types let url = window.clickTag || window.clickTag1 || window.clickTag2 || window.clickTag3 || window.clickTag4 || window.bsClickTAG || window.bsClickTAG1 || window.bsClickTAG2 || window.url || ''; if(typeof url == 'string'){ document.body.dataset['perxceptAdRedirectUrl'] = url;}.
Collapse
Affiliation(s)
| | | | | | - Umberto Malepelle
- Department of Public Health University Federico II of Naples, Naples, Italy
| | - Aakash Desai
- Division of Hematology and Oncology, Department of Medicine, University of Alabama, Birmingham, United States.
| |
Collapse
|
2
|
Haake SM, Rios BL, Pozzi A, Zent R. Integrating integrins with the hallmarks of cancer. Matrix Biol 2024; 130:20-35. [PMID: 38677444 DOI: 10.1016/j.matbio.2024.04.003] [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: 01/09/2024] [Revised: 04/02/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
Epithelial cells adhere to a specialized extracellular matrix called the basement membrane which allows them to polarize and form epithelial tissues. The extracellular matrix provides essential physical scaffolding and biochemical and biophysical cues required for tissue morphogenesis, differentiation, function, and homeostasis. Epithelial cell adhesion to the extracellular matrix (i.e., basement membrane) plays a critical role in organizing epithelial tissues, separating the epithelial cells from the stroma. Epithelial cell detachment from the basement membrane classically results in death, though detachment or invasion through the basement membrane represents a critical step in carcinogenesis. Epithelial cells bind to the extracellular matrix via specialized matrix receptors, including integrins. Integrins are transmembrane receptors that form a mechanical linkage between the extracellular matrix and the intracellular cytoskeleton and are required for anchorage-dependent cellular functions such as proliferation, migration, and invasion. The role of integrins in the development, growth, and dissemination of multiple types of carcinomas has been investigated by numerous methodologies, which has led to great complexity. To organize this vast array of information, we have utilized the "Hallmarks of Cancer" from Hanahan and Weinberg as a convenient framework to discuss the role of integrins in the pathogenesis of cancers. This review explores this biology and how its complexity has impacted the development of integrin-targeted anti-cancer therapeutics.
Collapse
Affiliation(s)
- Scott M Haake
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Veterans Affairs, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA; Cancer Biology Program, Vanderbilt University, Nashville, TN, USA.
| | - Brenda L Rios
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA; Cancer Biology Program, Vanderbilt University, Nashville, TN, USA
| | - Ambra Pozzi
- Department of Veterans Affairs, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Roy Zent
- Department of Veterans Affairs, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA; Cancer Biology Program, Vanderbilt University, Nashville, TN, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
3
|
Bamburowicz-Klimkowska M, Bystrzejewski M, Kasprzak A, Cieszanowski A, Grudzinski IP. Monoclonal antibody-navigated carbon-encapsulated iron nanoparticles used for MRI-based tracking integrin receptors in murine melanoma. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 55:102721. [PMID: 38007065 DOI: 10.1016/j.nano.2023.102721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/27/2023]
Abstract
Integrin beta-3 is a cell adhesion molecule that mediate cell-to-cell and cell-to-extracellular matrix communication. The major goal of this study was to explore melanoma cells (B16F10) based upon specific direct targeting of the β3 subunit (CD61) in the integrin αvβ3 receptor using carbon-encapsulated iron nanoparticles decorated with monoclonal antibodies (Fe@C-CONH-anti-CD61 and Fe@C-(CH2)2-CONH-anti-CD61). Both melanoma cells treated with nanoparticles as well as C57BL/6 mice bearing syngeneic B16-F10 tumors intravenously injected with nanoparticles were tested in preclinical MRI studies. The as-synthesized carbon-encapsulated iron nanoparticles functionalized with CD61 monoclonal antibodies have been successfully used as a novel targeted contrast agent for MRI-based tracking melanoma cells expressing the β3 subunit of the integrin αvβ3 receptor.
Collapse
Affiliation(s)
| | - Michal Bystrzejewski
- Department of Physical Chemistry, Faculty of Chemistry, Warsaw University, Pasteura 1, 02-093 Warsaw, Poland
| | - Artur Kasprzak
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Andrzej Cieszanowski
- Department of Clinical Radiology, Faculty of Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
| | - Ireneusz P Grudzinski
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| |
Collapse
|
4
|
Pang X, He X, Qiu Z, Zhang H, Xie R, Liu Z, Gu Y, Zhao N, Xiang Q, Cui Y. Targeting integrin pathways: mechanisms and advances in therapy. Signal Transduct Target Ther 2023; 8:1. [PMID: 36588107 PMCID: PMC9805914 DOI: 10.1038/s41392-022-01259-6] [Citation(s) in RCA: 433] [Impact Index Per Article: 216.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 01/03/2023] Open
Abstract
Integrins are considered the main cell-adhesion transmembrane receptors that play multifaceted roles as extracellular matrix (ECM)-cytoskeletal linkers and transducers in biochemical and mechanical signals between cells and their environment in a wide range of states in health and diseases. Integrin functions are dependable on a delicate balance between active and inactive status via multiple mechanisms, including protein-protein interactions, conformational changes, and trafficking. Due to their exposure on the cell surface and sensitivity to the molecular blockade, integrins have been investigated as pharmacological targets for nearly 40 years, but given the complexity of integrins and sometimes opposite characteristics, targeting integrin therapeutics has been a challenge. To date, only seven drugs targeting integrins have been successfully marketed, including abciximab, eptifibatide, tirofiban, natalizumab, vedolizumab, lifitegrast, and carotegrast. Currently, there are approximately 90 kinds of integrin-based therapeutic drugs or imaging agents in clinical studies, including small molecules, antibodies, synthetic mimic peptides, antibody-drug conjugates (ADCs), chimeric antigen receptor (CAR) T-cell therapy, imaging agents, etc. A serious lesson from past integrin drug discovery and research efforts is that successes rely on both a deep understanding of integrin-regulatory mechanisms and unmet clinical needs. Herein, we provide a systematic and complete review of all integrin family members and integrin-mediated downstream signal transduction to highlight ongoing efforts to develop new therapies/diagnoses from bench to clinic. In addition, we further discuss the trend of drug development, how to improve the success rate of clinical trials targeting integrin therapies, and the key points for clinical research, basic research, and translational research.
Collapse
Affiliation(s)
- Xiaocong Pang
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Xu He
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Zhiwei Qiu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Hanxu Zhang
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Ran Xie
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Zhiyan Liu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Yanlun Gu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Nan Zhao
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Qian Xiang
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034, Beijing, China. .,Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191, Beijing, China.
| | - Yimin Cui
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034, Beijing, China. .,Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191, Beijing, China.
| |
Collapse
|
5
|
Due SL, Watson DI, Bastian I, Eichelmann AK, Hussey DJ. Oestrogen Receptor Isoforms May Represent a Therapeutic Target in Oesophageal Adenocarcinoma. Cancers (Basel) 2022; 14:1891. [PMID: 35454796 PMCID: PMC9032750 DOI: 10.3390/cancers14081891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/18/2022] [Accepted: 03/26/2022] [Indexed: 02/01/2023] Open
Abstract
Oesophageal adenocarcinoma is a rapidly increasing problem in which treatment options are limited. Previous studies have shown that oesophageal adenocarcinoma cells and tissues express oestrogen receptors (ERs) and show growth suppression and apoptosis in response to ER modulator agents such as tamoxifen. ERs are known to be expressed in a number of isoforms that act together to regulate cell growth and cell death. In this study, we used western blotting to profile the expression of ERα and ERβ isoforms, and expression of the oncologically related molecules p53, HER2, and EGFR, in a panel of oesophageal adenocarcinoma cell lines. The cytotoxicity of tamoxifen in the cell lines was determined with Annexin V-FITC flow cytometry, and correlations between cytotoxicity and receptor expression were assessed using Spearman's rank-order correlation. Oesophageal adenocarcinoma cell lines showed varying cytotoxicity in response to tamoxifen. The ER species ERα90, ERα50, and ERα46, as well as p53, were positively associated with a cytotoxic response. Conversely, ERα74, ERα70, and ERβ54 were associated with a lack of cytotoxic response. The ER species detected in oesophageal adenocarcinoma cells may work together to confer sensitivity to ER modulators in this disease, which could open up a new avenue for therapy in selected patients.
Collapse
Affiliation(s)
- Steven L. Due
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA 5042, Australia; (S.L.D.); (D.I.W.)
- Flinders Health and Medical Research Institute—Cancer Program, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (I.B.); (A.-K.E.)
| | - David I. Watson
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA 5042, Australia; (S.L.D.); (D.I.W.)
- Flinders Health and Medical Research Institute—Cancer Program, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (I.B.); (A.-K.E.)
| | - Isabell Bastian
- Flinders Health and Medical Research Institute—Cancer Program, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (I.B.); (A.-K.E.)
| | - Ann-Kathrin Eichelmann
- Flinders Health and Medical Research Institute—Cancer Program, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (I.B.); (A.-K.E.)
| | - Damian J. Hussey
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA 5042, Australia; (S.L.D.); (D.I.W.)
- Flinders Health and Medical Research Institute—Cancer Program, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (I.B.); (A.-K.E.)
| |
Collapse
|
6
|
Li M, Wang Y, Li M, Wu X, Setrerrahmane S, Xu H. Integrins as attractive targets for cancer therapeutics. Acta Pharm Sin B 2021; 11:2726-2737. [PMID: 34589393 PMCID: PMC8463276 DOI: 10.1016/j.apsb.2021.01.004] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023] Open
Abstract
Integrins are transmembrane receptors that have been implicated in the biology of various human physiological and pathological processes. These molecules facilitate cell–extracellular matrix and cell–cell interactions, and they have been implicated in fibrosis, inflammation, thrombosis, and tumor metastasis. The role of integrins in tumor progression makes them promising targets for cancer treatment, and certain integrin antagonists, such as antibodies and synthetic peptides, have been effectively utilized in the clinic for cancer therapy. Here, we discuss the evidence and knowledge on the contribution of integrins to cancer biology. Furthermore, we summarize the clinical attempts targeting this family in anti-cancer therapy development.
Collapse
Key Words
- ADAMs, adisintegrin and metalloproteases
- AJ, adherens junctions
- Antagonists
- CAFs, cancer-associated fibroblasts
- CAR, chimeric antigen receptor
- CRC, colorectal cancer
- CSC, cancer stem cell
- Clinical trial
- ECM, extracellular matrix
- EGFR, epidermal growth factor receptor
- EMT, epithelial–mesenchymal transition
- ERK, extracellular regulated kinase
- Extracellular matrix
- FAK, focal adhesion kinase
- FDA, U.S. Food and Drug Administration
- HIF-1α, hypoxia-inducible factor-1α
- HUVECs, human umbilical vein endothelial cells
- ICAMs, intercellular adhesion molecules
- IGFR, insulin-like growth factor receptor
- IMD, integrin-mediated death
- Integrins
- JNK, c-Jun N-terminal kinase 16
- MAPK, mitogen-activated protein kinase
- MMP2, matrix metalloprotease 2
- NF-κB, nuclear factor-κB
- NSCLC, non-small cell lung cancer
- PDGFR, platelet-derived growth factor receptor
- PI3K, phosphatidylinositol 3-kinase
- RGD, Arg-Gly-Asp
- RTKs, receptor tyrosine kinases
- SAPKs, stress-activated MAP kinases
- SDF-1, stromal cell-derived factor-1
- SH2, Src homology 2
- STAT3, signal transducer and activator of transcription 3
- TCGA, The Cancer Genome Atlas
- TICs, tumor initiating cells
- TNF, tumor necrosis factor
- Targeted drug
- Tumor progression
- VCAMs, vascular cell adhesion molecules
- VEGFR, vascular endothelial growth factor receptor
- mAb, monoclonal antibodies
- sdCAR-T, switchable dual-receptor CAR-engineered T
- siRNA, small interference RNA
- uPA, urokinase-type plasminogen activator
Collapse
|
7
|
Dzobo K. Integrins Within the Tumor Microenvironment: Biological Functions, Importance for Molecular Targeting, and Cancer Therapeutics Innovation. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:417-430. [PMID: 34191612 DOI: 10.1089/omi.2021.0069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many cellular functions important for solid tumor initiation and progression are mediated by members of the integrin family, a diverse family of cell attachment receptors. With recent studies emphasizing the role of the tumor microenvironment (TME) in tumor initiation and progression, it is not surprising that considerable attention is being paid to integrins. Several integrin antagonists are under clinical trials, with many demonstrating promising activity in patients with different cancers. A deeper knowledge of the functions of integrins within the TME is still required and might lead to better inhibitors being discovered. Integrin expression is commonly dysregulated in many tumors with integrins playing key roles in signaling as well as promotion of tumor cell invasion and migration. Integrins also play a major role in adhesion of circulating tumor cells to new sites and the resulting formation of secondary tumors. Furthermore, integrins have demonstrated the ability to promoting stem cell-like properties in tumor cells as well as drug resistance. Anti-integrin therapies rely heavily on the doses or concentrations used as these determine whether the drugs act as antagonists or as integrin agonists. This expert review offers the latest synthesis in terms of the current knowledge of integrins functions within the TME and as potential molecular targets for cancer therapeutics innovation.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
8
|
El-Arabey AA, Denizli M, Kanlikilicer P, Bayraktar R, Ivan C, Rashed M, Kabil N, Ozpolat B, Calin GA, Salama SA, Abd-Allah AR, Sood AK, Lopez-Berestein G. GATA3 as a master regulator for interactions of tumor-associated macrophages with high-grade serous ovarian carcinoma. Cell Signal 2020; 68:109539. [PMID: 31935430 DOI: 10.1016/j.cellsig.2020.109539] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 02/03/2023]
Abstract
High-grade serous ovarian carcinoma (HGSOC) is the most lethal gynecologic cancer. Emerging evidence suggests that tumor-associated macrophages (TAMs) play an immunosuppressive role in the tumor microenvironment and promote tumor growth, angiogenesis, and metastasis in ovarian cancer. Therefore, targeting TAMs in patients with ovarian cancer is an appealing strategy; however, all trials to date have failed. To improve the efficacy of this approach, we sought to elucidate the underlying mechanisms of the role of TAMs in ovarian cancer. We found that the developmental transcription factor GATA3 was highly expressed in HGSOC cell lines but not in the fallopian tube, which is the main origin of HGSOC. GATA3 expression was associated with poor prognosis in HGSOC patients (P < .05) and was found to promote proliferation and migration in HGSOC cell lines. GATA3 was released abundantly from TAM cells via exosomes and contributed to tumor growth in the tumor microenvironment. Moreover, GATA3 acted as a regulator for macrophage polarization and interactions between TAMs and HGSOC to support proliferation, motility, and cisplatin chemoresistance in mutant TP53 HGSOC cell lines. Furthermore, GATA3 played a critical role in the interactions between TAMs and mutant TP53 HGSOC to promote angiogenesis and epithelial-mesenchymal transition with epigenetic regulation. Targeting GATA3 using GATA3siRNA in TAMs impeded GATA3-driven proliferation, migration, cisplatin chemoresistance, and angiogenesis in mutant TP53 HGSOC cell lines. Our findings indicate that GATA3 plays a novel role in immunoediting of HGSOC and demonstrate that GATA3 may serve as a prognostic marker for HGSOC and a promising target in the treatment of HGSOC.
Collapse
MESH Headings
- Apoptosis/genetics
- Cell Communication/genetics
- Cell Line, Tumor
- Cell Movement
- Cell Polarity/genetics
- Endometrial Neoplasms/pathology
- Endothelial Cells/pathology
- Epigenesis, Genetic
- Epithelial-Mesenchymal Transition/genetics
- Exosomes/metabolism
- Exosomes/ultrastructure
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Female
- GATA3 Transcription Factor/metabolism
- Genome, Human
- Humans
- Matrix Metalloproteinase 9/metabolism
- Mutation/genetics
- Neoplasm Grading
- Neoplasm Proteins/metabolism
- Neoplasms, Cystic, Mucinous, and Serous/genetics
- Neoplasms, Cystic, Mucinous, and Serous/metabolism
- Neoplasms, Cystic, Mucinous, and Serous/pathology
- Neovascularization, Pathologic/genetics
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Phosphorylation
- RNA Splice Sites/genetics
- Tumor Microenvironment/genetics
- Tumor Suppressor Protein p53/genetics
- Tumor-Associated Macrophages/metabolism
- Tumor-Associated Macrophages/pathology
Collapse
Affiliation(s)
- Amr Ahmed El-Arabey
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Pharmacology and Toxicology, Al-Azhar University, Faculty of Pharmacy, Cairo, Egypt
| | - Merve Denizli
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pinar Kanlikilicer
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Recep Bayraktar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mohammed Rashed
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Pharmacology and Toxicology, Al-Azhar University, Faculty of Pharmacy, Cairo, Egypt
| | - Nashwa Kabil
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Salama Abdou Salama
- Department of Pharmacology and Toxicology, Al-Azhar University, Faculty of Pharmacy, Cairo, Egypt
| | - Adel Rashad Abd-Allah
- Department of Pharmacology and Toxicology, Al-Azhar University, Faculty of Pharmacy, Cairo, Egypt
| | - Anil K Sood
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
9
|
Gallo E, Kelil A, Bayliss PE, Jeganathan A, Egorova O, Ploder L, Adams JJ, Giblin P, Sidhu SS. In situ antibody phage display yields optimal inhibitors of integrin α11/β1. MAbs 2020; 12:1717265. [PMID: 31980006 PMCID: PMC6999838 DOI: 10.1080/19420862.2020.1717265] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/30/2019] [Accepted: 01/09/2020] [Indexed: 01/13/2023] Open
Abstract
Integrins are transmembrane multi-conformation receptors that mediate interactions with the extracellular matrix. In cancer, integrins influence metastasis, proliferation, and survival. Collagen-binding integrin-α11/β1, a marker of aggressive tumors that is involved in stroma-tumor crosstalk, may be an attractive target for anti-cancer therapeutic antibodies. We performed selections with phage-displayed synthetic antibody libraries for binding to either purified integrin-α11/β1 or in situ on live cells. The in-situ strategy yielded many diverse antibodies, and strikingly, most of these antibodies did not recognize purified integrin-α11/β1. Conversely, none of the antibodies selected for binding to purified integrin-α11/β1 were able to efficiently recognize native cell-surface antigen. Most importantly, only the in-situ selection yielded functional antibodies that were able to compete with collagen-I for binding to cell-surface integrin-α11/β1, and thus inhibited cell adhesion. In-depth characterization of a subset of in situ-derived clones as full-length immunoglobulins revealed high affinity cellular binding and inhibitory activities in the single-digit nanomolar range. Moreover, the antibodies showed high selectivity for integrin-α11/β1 with minimal cross-reactivity for close homologs. Taken together, our findings highlight the advantages of in-situ selections for generation of anti-integrin antibodies optimized for recognition and inhibition of native cell-surface proteins, and our work establishes general methods that could be extended to many other membrane proteins.
Collapse
Affiliation(s)
- Eugenio Gallo
- Department of Molecular Genetics, University of Toronto, Donnelly Centre, Toronto, Ontario, Canada
| | - Abdellali Kelil
- Department of Molecular Genetics, University of Toronto, Donnelly Centre, Toronto, Ontario, Canada
| | - Peter E. Bayliss
- Northern Biologics, Inc., Princess Margaret Cancer Research Tower, Toronto, Ontario, Canada
| | - Ajitha Jeganathan
- Northern Biologics, Inc., Princess Margaret Cancer Research Tower, Toronto, Ontario, Canada
| | - Olga Egorova
- Northern Biologics, Inc., Princess Margaret Cancer Research Tower, Toronto, Ontario, Canada
| | - Lynda Ploder
- Department of Molecular Genetics, University of Toronto, Donnelly Centre, Toronto, Ontario, Canada
| | - Jarret J. Adams
- Department of Molecular Genetics, University of Toronto, Donnelly Centre, Toronto, Ontario, Canada
| | - Patricia Giblin
- Northern Biologics, Inc., Princess Margaret Cancer Research Tower, Toronto, Ontario, Canada
| | - Sachdev S. Sidhu
- Department of Molecular Genetics, University of Toronto, Donnelly Centre, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Smirnov A, Cappello A, Lena AM, Anemona L, Mauriello A, Di Daniele N, Annicchiarico-Petruzzelli M, Melino G, Candi E. ZNF185 is a p53 target gene following DNA damage. Aging (Albany NY) 2019; 10:3308-3326. [PMID: 30446632 PMCID: PMC6286825 DOI: 10.18632/aging.101639] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/01/2018] [Indexed: 12/17/2022]
Abstract
The transcription factor p53 is a key player in the tumour suppressive DNA damage response and a growing number of target genes involved in these pathways has been identified. p53 has been shown to be implicated in controlling cell motility and its mutant form enhances metastasis by loss of cell directionality, but the p53 role in this context has not yet being investigated. Here, we report that ZNF185, an actin cytoskeleton-associated protein from LIM-family of Zn-finger proteins, is induced following DNA-damage. ChIP-seq analysis, chromatin crosslinking immune-precipitation experiments and luciferase assays demonstrate that ZNF185 is a bona fide p53 target gene. Upon genotoxic stress, caused by DNA-damaging drug etoposide and UVB irradiation, ZNF185 expression is up-regulated and in etoposide-treated cells, ZNF185 depletion does not affect cell proliferation and apoptosis, but interferes with actin cytoskeleton remodelling and cell polarization. Bioinformatic analysis of different types of epithelial cancers from both TCGA and GTEx databases showed a significant decrease in ZNF185 mRNA level compared to normal tissues. These findings are confirmed by tissue micro-array IHC staining. Our data highlight the involvement of ZNF185 and cytoskeleton changes in p53-mediated cellular response to genotoxic stress and indicate ZNF185 as potential biomarker for epithelial cancer diagnosis.
Collapse
Affiliation(s)
- Artem Smirnov
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Angela Cappello
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Anna Maria Lena
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Lucia Anemona
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Nicola Di Daniele
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome 00133, Italy
| | | | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy.,MRC-Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy.,Istituto Dermopatico dell'Immacolata-IRCCS, Rome 00163, Italy
| |
Collapse
|
11
|
Harjunpää H, Llort Asens M, Guenther C, Fagerholm SC. Cell Adhesion Molecules and Their Roles and Regulation in the Immune and Tumor Microenvironment. Front Immunol 2019; 10:1078. [PMID: 31231358 PMCID: PMC6558418 DOI: 10.3389/fimmu.2019.01078] [Citation(s) in RCA: 481] [Impact Index Per Article: 80.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/29/2019] [Indexed: 12/14/2022] Open
Abstract
The immune system and cancer have a complex relationship with the immune system playing a dual role in tumor development. The effector cells of the immune system can recognize and kill malignant cells while immune system-mediated inflammation can also promote tumor growth and regulatory cells suppress the anti-tumor responses. In the center of all anti-tumor responses is the ability of the immune cells to migrate to the tumor site and to interact with each other and with the malignant cells. Cell adhesion molecules including receptors of the immunoglobulin superfamily and integrins are of crucial importance in mediating these processes. Particularly integrins play a vital role in regulating all aspects of immune cell function including immune cell trafficking into tissues, effector cell activation and proliferation and the formation of the immunological synapse between immune cells or between immune cell and the target cell both during homeostasis and during inflammation and cancer. In this review we discuss the molecular mechanisms regulating integrin function and the role of integrins and other cell adhesion molecules in immune responses and in the tumor microenvironment. We also describe how malignant cells can utilize cell adhesion molecules to promote tumor growth and metastases and how these molecules could be targeted in cancer immunotherapy.
Collapse
Affiliation(s)
- Heidi Harjunpää
- Research Program of Molecular and Integrative Biosciences, Faculty of Bio- and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Marc Llort Asens
- Research Program of Molecular and Integrative Biosciences, Faculty of Bio- and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Carla Guenther
- Research Program of Molecular and Integrative Biosciences, Faculty of Bio- and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Susanna C Fagerholm
- Research Program of Molecular and Integrative Biosciences, Faculty of Bio- and Environmental Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
12
|
Parks A, Marceau F. Lysosomotropic cationic drugs induce cytostatic and cytotoxic effects: Role of liposolubility and autophagic flux and antagonism by cholesterol ablation. Toxicol Appl Pharmacol 2016; 305:55-65. [DOI: 10.1016/j.taap.2016.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/03/2016] [Accepted: 06/07/2016] [Indexed: 12/16/2022]
|
13
|
Pauty J, Côté MF, Rodrigue A, Velic D, Masson JY, Fortin S. Investigation of the DNA damage response to SFOM-0046, a new small-molecule drug inducing DNA double-strand breaks. Sci Rep 2016; 6:23302. [PMID: 27001483 PMCID: PMC4802344 DOI: 10.1038/srep23302] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 03/03/2016] [Indexed: 01/25/2023] Open
Abstract
2-Ethylphenyl 4-(3-ethylureido)benzenesulfonate (SFOM-0046) is a novel anticancer agent that arrests cell cycle in S-phase and causes DNA replication stress leading to the phosphorylation of H2AX into γ-H2AX. First, using the M21, HT29, HT-1080 and HeLa cell lines, we confirmed that S-phase cell cycle arrest and γ-H2AX foci induction by SFOM-0046 is a general mechanism occurring in diverse cancer cell lines. In addition to γ-H2AX, SFOM-0046 activates preferentially ATR-Chk1 in M21 and HT29 cells while both ATR-Chk1 and ATM-Chk2 pathways are activated in HCT116 cells. Co-localization of SFOM-0046-induced 53BP1 foci with γ-H2AX foci validates that the DNA damage generated corresponds to double-strand-breaks (DSBs). Consistent with an S-phase arrest, SFOM-0046 treatment induces RAD51 foci formation but not DNA-PKcs foci, confirming that homologous recombination is the major DSB repair pathway targeted by the drug. Furthermore, using isogenic HCT116 p53+/+ and HCT116 p53−/− cells, we showed that p53 plays a key role in the survival mechanism to SFOM-0046. Finally, SFOM-0046 exhibits a dose-dependent antitumor activity on human fibrosarcoma HT-1080 tumours grafted onto chick chorioallantoic membranes without showing embryo toxicity even at high doses. Altogether, our results highlight SFOM-0046 as a very promising drug that induces a replication stress response.
Collapse
Affiliation(s)
- Joris Pauty
- Genome Stability Laboratory, CHU de Québec Research Center, Oncology Axis, Hôtel-Dieu-de-Québec, 9 McMahon, Quebec City, QC, G1R 2J6, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Quebec City, QC, G1V 0A6, Canada
| | - Marie-France Côté
- CHU de Québec Research Center, Oncology Axis, Hôpital Saint-François d'Assise, 10 de l'Espinay, Quebec City, QC, G1L 3L5, Canada
| | - Amélie Rodrigue
- Genome Stability Laboratory, CHU de Québec Research Center, Oncology Axis, Hôtel-Dieu-de-Québec, 9 McMahon, Quebec City, QC, G1R 2J6, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Quebec City, QC, G1V 0A6, Canada
| | - Denis Velic
- Genome Stability Laboratory, CHU de Québec Research Center, Oncology Axis, Hôtel-Dieu-de-Québec, 9 McMahon, Quebec City, QC, G1R 2J6, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Quebec City, QC, G1V 0A6, Canada
| | - Jean-Yves Masson
- Genome Stability Laboratory, CHU de Québec Research Center, Oncology Axis, Hôtel-Dieu-de-Québec, 9 McMahon, Quebec City, QC, G1R 2J6, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Quebec City, QC, G1V 0A6, Canada
| | - Sébastien Fortin
- CHU de Québec Research Center, Oncology Axis, Hôpital Saint-François d'Assise, 10 de l'Espinay, Quebec City, QC, G1L 3L5, Canada.,Faculty of Pharmacy, Laval University, Quebec City, QC, G1V 0A6, Canada
| |
Collapse
|
14
|
Meves A, Nikolova E, Heim JB, Squirewell EJ, Cappel MA, Pittelkow MR, Otley CC, Behrendt N, Saunte DM, Lock-Andersen J, Schenck LA, Weaver AL, Suman VJ. Tumor Cell Adhesion As a Risk Factor for Sentinel Lymph Node Metastasis in Primary Cutaneous Melanoma. J Clin Oncol 2015; 33:2509-15. [PMID: 26150443 DOI: 10.1200/jco.2014.60.7002] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Less than 20% of patients with melanoma who undergo sentinel lymph node (SLN) biopsy based on American Society of Clinical Oncology/Society of Surgical Oncology recommendations are SLN positive. We present a multi-institutional study to discover new molecular risk factors associated with SLN positivity in thin and intermediate-thickness melanoma. PATIENTS AND METHODS Gene clusters with functional roles in melanoma metastasis were discovered by next-generation sequencing and validated by quantitative polymerase chain reaction using a discovery set of 73 benign nevi, 76 primary cutaneous melanoma, and 11 in-transit melanoma metastases. We then used polymerase chain reaction to quantify gene expression in a model development cohort of 360 consecutive thin and intermediate-thickness melanomas and a validation cohort of 146 melanomas. Outcome of interest was SLN biopsy metastasis within 90 days of melanoma diagnosis. Logic and logistic regression analyses were used to develop a model for the likelihood of SLN metastasis from molecular, clinical, and histologic variables. RESULTS ITGB3, LAMB1, PLAT, and TP53 expression were associated with SLN metastasis. The predictive ability of a model that included these molecular variables in combination with clinicopathologic variables (patient age, Breslow depth, and tumor ulceration) was significantly greater than a model that only considered clinicopathologic variables and also performed well in the validation cohort (area under the curve, 0.93; 95% CI, 0.87 to 0.97; false-positive and false-negative rates of 22% and 0%, respectively, using a 10% cutoff for predicted SLN metastasis risk). CONCLUSION The addition of cell adhesion-linked gene expression variables to clinicopathologic variables improves the identification of patients with SLN metastases within 90 days of melanoma diagnosis.
Collapse
Affiliation(s)
- Alexander Meves
- Alexander Meves, Ekaterina Nikolova, Joel B. Heim, Edwin J. Squirewell, Clark C. Otley, Louis A. Schenck, Amy L. Weaver, and Vera J. Suman, Mayo Clinic, Rochester, MN; Mark A. Cappel, Mayo Clinic, Jacksonville, FL; Mark R. Pittelkow, Mayo Clinic, Scottsdale, AZ; and Nille Behrendt, Ditte M. Saunte, and Jorgen Lock-Andersen, Hospital Roskilde, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Ekaterina Nikolova
- Alexander Meves, Ekaterina Nikolova, Joel B. Heim, Edwin J. Squirewell, Clark C. Otley, Louis A. Schenck, Amy L. Weaver, and Vera J. Suman, Mayo Clinic, Rochester, MN; Mark A. Cappel, Mayo Clinic, Jacksonville, FL; Mark R. Pittelkow, Mayo Clinic, Scottsdale, AZ; and Nille Behrendt, Ditte M. Saunte, and Jorgen Lock-Andersen, Hospital Roskilde, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Joel B Heim
- Alexander Meves, Ekaterina Nikolova, Joel B. Heim, Edwin J. Squirewell, Clark C. Otley, Louis A. Schenck, Amy L. Weaver, and Vera J. Suman, Mayo Clinic, Rochester, MN; Mark A. Cappel, Mayo Clinic, Jacksonville, FL; Mark R. Pittelkow, Mayo Clinic, Scottsdale, AZ; and Nille Behrendt, Ditte M. Saunte, and Jorgen Lock-Andersen, Hospital Roskilde, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Edwin J Squirewell
- Alexander Meves, Ekaterina Nikolova, Joel B. Heim, Edwin J. Squirewell, Clark C. Otley, Louis A. Schenck, Amy L. Weaver, and Vera J. Suman, Mayo Clinic, Rochester, MN; Mark A. Cappel, Mayo Clinic, Jacksonville, FL; Mark R. Pittelkow, Mayo Clinic, Scottsdale, AZ; and Nille Behrendt, Ditte M. Saunte, and Jorgen Lock-Andersen, Hospital Roskilde, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mark A Cappel
- Alexander Meves, Ekaterina Nikolova, Joel B. Heim, Edwin J. Squirewell, Clark C. Otley, Louis A. Schenck, Amy L. Weaver, and Vera J. Suman, Mayo Clinic, Rochester, MN; Mark A. Cappel, Mayo Clinic, Jacksonville, FL; Mark R. Pittelkow, Mayo Clinic, Scottsdale, AZ; and Nille Behrendt, Ditte M. Saunte, and Jorgen Lock-Andersen, Hospital Roskilde, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mark R Pittelkow
- Alexander Meves, Ekaterina Nikolova, Joel B. Heim, Edwin J. Squirewell, Clark C. Otley, Louis A. Schenck, Amy L. Weaver, and Vera J. Suman, Mayo Clinic, Rochester, MN; Mark A. Cappel, Mayo Clinic, Jacksonville, FL; Mark R. Pittelkow, Mayo Clinic, Scottsdale, AZ; and Nille Behrendt, Ditte M. Saunte, and Jorgen Lock-Andersen, Hospital Roskilde, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Clark C Otley
- Alexander Meves, Ekaterina Nikolova, Joel B. Heim, Edwin J. Squirewell, Clark C. Otley, Louis A. Schenck, Amy L. Weaver, and Vera J. Suman, Mayo Clinic, Rochester, MN; Mark A. Cappel, Mayo Clinic, Jacksonville, FL; Mark R. Pittelkow, Mayo Clinic, Scottsdale, AZ; and Nille Behrendt, Ditte M. Saunte, and Jorgen Lock-Andersen, Hospital Roskilde, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nille Behrendt
- Alexander Meves, Ekaterina Nikolova, Joel B. Heim, Edwin J. Squirewell, Clark C. Otley, Louis A. Schenck, Amy L. Weaver, and Vera J. Suman, Mayo Clinic, Rochester, MN; Mark A. Cappel, Mayo Clinic, Jacksonville, FL; Mark R. Pittelkow, Mayo Clinic, Scottsdale, AZ; and Nille Behrendt, Ditte M. Saunte, and Jorgen Lock-Andersen, Hospital Roskilde, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ditte M Saunte
- Alexander Meves, Ekaterina Nikolova, Joel B. Heim, Edwin J. Squirewell, Clark C. Otley, Louis A. Schenck, Amy L. Weaver, and Vera J. Suman, Mayo Clinic, Rochester, MN; Mark A. Cappel, Mayo Clinic, Jacksonville, FL; Mark R. Pittelkow, Mayo Clinic, Scottsdale, AZ; and Nille Behrendt, Ditte M. Saunte, and Jorgen Lock-Andersen, Hospital Roskilde, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jorgen Lock-Andersen
- Alexander Meves, Ekaterina Nikolova, Joel B. Heim, Edwin J. Squirewell, Clark C. Otley, Louis A. Schenck, Amy L. Weaver, and Vera J. Suman, Mayo Clinic, Rochester, MN; Mark A. Cappel, Mayo Clinic, Jacksonville, FL; Mark R. Pittelkow, Mayo Clinic, Scottsdale, AZ; and Nille Behrendt, Ditte M. Saunte, and Jorgen Lock-Andersen, Hospital Roskilde, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Louis A Schenck
- Alexander Meves, Ekaterina Nikolova, Joel B. Heim, Edwin J. Squirewell, Clark C. Otley, Louis A. Schenck, Amy L. Weaver, and Vera J. Suman, Mayo Clinic, Rochester, MN; Mark A. Cappel, Mayo Clinic, Jacksonville, FL; Mark R. Pittelkow, Mayo Clinic, Scottsdale, AZ; and Nille Behrendt, Ditte M. Saunte, and Jorgen Lock-Andersen, Hospital Roskilde, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amy L Weaver
- Alexander Meves, Ekaterina Nikolova, Joel B. Heim, Edwin J. Squirewell, Clark C. Otley, Louis A. Schenck, Amy L. Weaver, and Vera J. Suman, Mayo Clinic, Rochester, MN; Mark A. Cappel, Mayo Clinic, Jacksonville, FL; Mark R. Pittelkow, Mayo Clinic, Scottsdale, AZ; and Nille Behrendt, Ditte M. Saunte, and Jorgen Lock-Andersen, Hospital Roskilde, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vera J Suman
- Alexander Meves, Ekaterina Nikolova, Joel B. Heim, Edwin J. Squirewell, Clark C. Otley, Louis A. Schenck, Amy L. Weaver, and Vera J. Suman, Mayo Clinic, Rochester, MN; Mark A. Cappel, Mayo Clinic, Jacksonville, FL; Mark R. Pittelkow, Mayo Clinic, Scottsdale, AZ; and Nille Behrendt, Ditte M. Saunte, and Jorgen Lock-Andersen, Hospital Roskilde, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
PICKARSKI MAUREEN, GLEASON ALEXA, BEDNAR BOHUMIL, DUONG LET. Orally active αvβ3 integrin inhibitor MK-0429 reduces melanoma metastasis. Oncol Rep 2015; 33:2737-45. [PMID: 25872534 PMCID: PMC4431436 DOI: 10.3892/or.2015.3910] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 03/20/2015] [Indexed: 11/16/2022] Open
Abstract
Melanoma remains one of the most aggressive types of cancer with a historically low survival rate. The αvβ3 integrin is involved in the progression of malignant melanoma. In the present study, the efficacy of MK-0429, a selective inhibitor of the αvβ3 integrin, was evaluated for its potential in the prevention of melanoma metastasis. Female B6D2F1 mice injected via the tail vein with murine B16F10 melanoma developed lung metastases within ~10 days. In the first experiment, the prevention of lung metastasis was assessed in the model treated with either vehicle, MK-0429 at 100 and 300 mg/kg orally twice daily or cyclophosphamide at 300 mg/kg, i.p. once daily. Study endpoints included determination of the study time period to achieve metastasis in lungs in this model, evaluation of the health effects on the study animals, the total number of lung colonies identified and lung tumor area. Unlike cyclophosphamide, the MK-0429 treatment did not lead to a significant weight reduction in mice. MK-0429 at 100 and 300 mg/kg reduced the number of metastatic tumor colonies by 64 and 57%, respectively, and the high dose also reduced the tumor area by 60% as compared to the vehicle. The second experiment employed B16F10 luciferase-expressing cells to examine the de novo progression of melanoma metastasis over 15 days with bioluminescent imaging of mice treated with MK-0429 at 300 mg/kg as compared to the vehicle. Tumor burden progressively advanced in the lungs of the B16F10-treated animals. However, MK-0429 reduced the progression of ventral and dorsal lung metastases by 22 and 38%, respectively, as compared to the vehicle, by study completion. Quantification of ex vivo tumor burden showed a 30-40% reduction in lung colonies by MK-0429. The two studies collectively demonstrated that MK-0429 was safe and efficacious in significantly decreasing melanoma metastasis in the lungs. The results emphasized the potential of MK-0429 as a novel, therapeutic agent for the prevention of metastatic melanoma.
Collapse
Affiliation(s)
| | - ALEXA GLEASON
- Imaging, Merck Research Laboratories, West Point, PA 19486, USA
| | - BOHUMIL BEDNAR
- Imaging, Merck Research Laboratories, West Point, PA 19486, USA
| | | |
Collapse
|
16
|
A membrane-type-1 matrix metalloproteinase (MT1-MMP)-discoidin domain receptor 1 axis regulates collagen-induced apoptosis in breast cancer cells. PLoS One 2015; 10:e0116006. [PMID: 25774665 PMCID: PMC4638154 DOI: 10.1371/journal.pone.0116006] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 12/03/2014] [Indexed: 11/19/2022] Open
Abstract
During tumour dissemination, invading breast carcinoma cells become confronted with a reactive stroma, a type I collagen-rich environment endowed with anti-proliferative and pro-apoptotic properties. To develop metastatic capabilities, tumour cells must acquire the capacity to cope with this novel microenvironment. How cells interact with and respond to their microenvironment during cancer dissemination remains poorly understood. To address the impact of type I collagen on the fate of tumour cells, human breast carcinoma MCF-7 cells were cultured within three-dimensional type I collagen gels (3D COL1). Using this experimental model, we have previously demonstrated that membrane type-1 matrix metalloproteinase (MT1-MMP), a proteinase overexpressed in many aggressive tumours, promotes tumour progression by circumventing the collagen-induced up-regulation of BIK, a pro-apoptotic tumour suppressor, and hence apoptosis. Here we performed a transcriptomic analysis to decipher the molecular mechanisms regulating 3D COL1-induced apoptosis in human breast cancer cells. Control and MT1-MMP expressing MCF-7 cells were cultured on two-dimensional plastic plates or within 3D COL1 and a global transcriptional time-course analysis was performed. Shifting the cells from plastic plates to 3D COL1 activated a complex reprogramming of genes implicated in various biological processes. Bioinformatic analysis revealed a 3D COL1-mediated alteration of key cellular functions including apoptosis, cell proliferation, RNA processing and cytoskeleton remodelling. By using a panel of pharmacological inhibitors, we identified discoidin domain receptor 1 (DDR1), a receptor tyrosine kinase specifically activated by collagen, as the initiator of 3D COL1-induced apoptosis. Our data support the concept that MT1-MMP contributes to the inactivation of the DDR1-BIK signalling axis through the cleavage of collagen fibres and/or the alteration of DDR1 receptor signalling unit, without triggering a drastic remodelling of the transcriptome of MCF-7 cells.
Collapse
|
17
|
Savar A, Acin S, Gonzalez CL, El-Sawy T, Mejia O, Li Z, Esmaeli B, Lacy-Hulbert A, El-Naggar A, McCarty JH, Caulin C. Loss of epithelial p53 and αv integrin cooperate through Akt to induce squamous cell carcinoma yet prevent remodeling of the tumor microenvironment. Oncogene 2015; 34:516-24. [PMID: 24469034 PMCID: PMC4303549 DOI: 10.1038/onc.2013.585] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 11/26/2013] [Accepted: 12/07/2013] [Indexed: 12/12/2022]
Abstract
Most of the squamous cell carcinomas (SCCs) of the skin and head and neck contain p53 mutations. The presence of p53 mutations in premalignant lesions suggests that they represent early events during tumor progression and additional alterations may be required for SCC development. Here we show that codeletion of the p53 and αv integrin genes in mouse stratified epithelia induced SCCs in 100% of the mice, more frequently and with much shorter latency than deletion of either gene alone. The SCCs that lacked p53 and αv in the epithelial tumor cells exhibited high Akt activity, lacked multiple types of infiltrating immune cells, contained a defective vasculature and grew slower than tumors that expressed p53 or αv. These results reveal that loss of αv in epithelial cells that lack p53 promotes SCC development, but also prevents remodeling of the tumor microenvironment and delays tumor growth. We observed that Akt inactivation in SCC cells that lack p53 and αv promoted anoikis. Thus, tumors may arise in these mice as a result of the increased cell survival induced by Akt activation triggered by loss of αv and p53, and by the defective recruitment of immune cells to these tumors, which may allow immune evasion. However, the defective vasculature and lack of a supportive stroma create a restrictive microenvironment in these SCCs that slows their growth. These mechanisms may underlie the rapid onset and slow growth of SCCs that lack p53 and αv.
Collapse
Affiliation(s)
- Aaron Savar
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sergio Acin
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cassandra L. Gonzalez
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tarek El-Sawy
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Olga Mejia
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhongyou Li
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bita Esmaeli
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Adam Lacy-Hulbert
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Adel El-Naggar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joseph H. McCarty
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Carlos Caulin
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
18
|
Arruda Macêdo JK, Fox JW, de Souza Castro M. Disintegrins from snake venoms and their applications in cancer research and therapy. Curr Protein Pept Sci 2015; 16:532-48. [PMID: 26031306 PMCID: PMC4997955 DOI: 10.2174/1389203716666150515125002] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 04/17/2015] [Accepted: 05/13/2015] [Indexed: 01/01/2023]
Abstract
Integrins regulate diverse functions in cancer pathology and in tumor cell development and contribute to important processes such as cell shape, survival, proliferation, transcription, angiogenesis, migration, and invasion. A number of snake venom proteins have the ability to interact with integrins. Among these are the disintegrins, a family of small, non-enzymatic, and cysteine-rich proteins found in the venom of numerous snake families. The venom proteins may have a potential role in terms of novel therapeutic leads for cancer treatment. Disintegrin can target specific integrins and as such it is conceivable that they could interfere in important processes involved in carcinogenesis, tumor growth, invasion and migration. Herein we present a survey of studies involving the use of snake venom disintegrins for cancer detection and treatment. The aim of this review is to highlight the relationship of integrins with cancer and to present examples as to how certain disintegrins can detect and affect biological processes related to cancer. This in turn will illustrate the great potential of these molecules for cancer research. Furthermore, we also outline several new approaches being created to address problems commonly associated with the clinical application of peptide-based drugs such as instability, immunogenicity, and availability.
Collapse
Affiliation(s)
| | - Jay W Fox
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, USA.
| | | |
Collapse
|
19
|
Liu P, Feng J, Kong F, Lu Q, Xu H, Meng J, Jiang Y. Gax inhibits perivascular preadipocyte biofunction mediated by IGF-1 induced FAK/Pyk2 and ERK2 cooperative pathways. Cell Signal 2014; 26:3036-45. [DOI: 10.1016/j.cellsig.2014.09.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/25/2014] [Indexed: 02/06/2023]
|
20
|
Inhibition of tumor-associated αvβ3 integrin regulates the angiogenic switch by enhancing expression of IGFBP-4 leading to reduced melanoma growth and angiogenesis in vivo. Angiogenesis 2014; 18:31-46. [PMID: 25249331 DOI: 10.1007/s10456-014-9445-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 09/16/2014] [Indexed: 12/20/2022]
Abstract
A more complete understanding of the mechanisms that regulate the angiogenic switch, which contributes to the conversion of small dormant tumors to actively growing malignancies, is important for the development of more effective anti-angiogenic strategies for cancer therapy. While significant progress has been made in understanding the complex mechanisms by which integrin αvβ3 expressed in endothelial cells governs angiogenesis, less is known concerning the ability of αvβ3 expressed within the tumor cell compartment to modulate the angiogenic output of a tumor. Here we provide evidence that αvβ3 expressed in melanoma cells may contribute to the suppression of IGFBP-4, an important negative regulator of IGF-1 signaling. Given the multiple context-dependent roles for αvβ3 in angiogenesis and tumor progression, our novel findings provide additional molecular insight into how αvβ3 may govern the angiogenic switch by a mechanism associated with a p38 MAPK and matrix metalloproteinases-dependent regulation of the endogenous angiogenesis inhibitor IGFBP-4.
Collapse
|
21
|
Bai SY, Xu N, Chen C, Song Y, Hu J, Bai C. Integrin αvβ5 as a biomarker for the assessment of non‐small cell lung cancer metastasis and overall survival. CLINICAL RESPIRATORY JOURNAL 2014; 9:457-67. [PMID: 24815623 DOI: 10.1111/crj.12163] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 05/01/2014] [Accepted: 05/08/2014] [Indexed: 01/12/2023]
Affiliation(s)
- Sally Yan Bai
- Department of Pulmonary Medicine Zhongshan Hospital Fudan University Shanghai China
| | - Nuo Xu
- Department of Pulmonary Medicine Zhongshan Hospital Fudan University Shanghai China
| | - Cuicui Chen
- Department of Pulmonary Medicine Zhongshan Hospital Fudan University Shanghai China
| | - Yuan‐lin Song
- Department of Pulmonary Medicine Zhongshan Hospital Fudan University Shanghai China
| | - Jie Hu
- Department of Pulmonary Medicine Zhongshan Hospital Fudan University Shanghai China
| | - Chun‐xue Bai
- Department of Pulmonary Medicine Zhongshan Hospital Fudan University Shanghai China
| |
Collapse
|
22
|
Schlesinger M, Roblek M, Ortmann K, Naggi A, Torri G, Borsig L, Bendas G. The role of VLA-4 binding for experimental melanoma metastasis and its inhibition by heparin. Thromb Res 2014; 133:855-62. [DOI: 10.1016/j.thromres.2014.02.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/31/2014] [Accepted: 02/21/2014] [Indexed: 12/31/2022]
|
23
|
Duperret EK, Ridky TW. Focal adhesion complex proteins in epidermis and squamous cell carcinoma. Cell Cycle 2013; 12:3272-85. [PMID: 24036537 DOI: 10.4161/cc.26385] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Focal adhesions (FAs) are large, integrin-containing, multi-protein assemblies spanning the plasma membrane that link the cellular cytoskeleton to surrounding extracellular matrix. They play critical roles in adhesion and cell signaling and are major regulators of epithelial homeostasis, tissue response to injury, and tumorigenesis. Most integrin subunits and their associated FA proteins are expressed in skin, and murine genetic models have provided insight into the functional roles of FAs in normal and neoplastic epidermis. Here, we discuss the roles of these proteins in normal epidermal proliferation, adhesion, wound healing, and cancer. While many downstream signaling mechanisms remain unclear, the critically important roles of FAs are highlighted by the development of therapeutics targeting FAs for human cancer.
Collapse
|
24
|
Smalley KS, Lioni M, Noma K, Haass NK, Herlyn M. In vitro three-dimensional tumor microenvironment models for anticancer drug discovery. Expert Opin Drug Discov 2013; 3:1-10. [PMID: 23480136 DOI: 10.1517/17460441.3.1.1] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Anticancer drug discovery has long been hampered by the poor predictivity of the preclinical models. There is a growing realization that the tumor microenvironment is a critical determinant of the response of cancer cells to therapeutic agents. The past 5 years have seen a great deal of progress in our understanding of how the three-dimensional microenvironment modulates the signaling behavior of tumor cells. The present review discusses how three-dimensional in vitro cell culture models can benefit cancer drug discovery through an accurate modeling of the tumor microenvironment, leading to more physiologically relevant experimental outcomes.
Collapse
Affiliation(s)
- Keiran Sm Smalley
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA +1 215 898 0002 ;
| | | | | | | | | |
Collapse
|
25
|
Abstract
The tumor associated stroma has been described in recent years as being complicit in tumor growth in pancreatic cancer. The stroma hosts a variety of components of both cellular and molecular makeup. In normal tissues, the stroma provides nutrients and regulatory signals for proper cellular polarity and function. However, following oncogenic transformation, the stromal compartment is conscripted to provide stimulatory signals and protection to tumor cells. It is these tumor-stromal interactions that are currently of great therapeutic interest. Several key reports have suggested that therapeutic targeting of the tumor-stromal interactions in pancreatic cancer has the potential to offer survival benefit. In this review, we will discuss the tumor-stromal interactions that contribute to tumor growth and progression, and ways in which we might counter these interactions.
Collapse
Affiliation(s)
- Clifford Whatcott
- Clinical Translational Research Division, The Translational Genomics Research Institute (TGEN), Phoenix, Arizona 85004, USA.
| | | | | | | |
Collapse
|
26
|
Hodjat M, Haller H, Dumler I, Kiyan Y. Urokinase receptor mediates doxorubicin-induced vascular smooth muscle cell senescence via proteasomal degradation of TRF2. J Vasc Res 2012; 50:109-23. [PMID: 23172421 DOI: 10.1159/000343000] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 08/23/2012] [Indexed: 11/19/2022] Open
Abstract
The anthracycline doxorubicin is a widely used effective anti-cancer drug. However, its application and dosage are severely limited due to its cardiotoxicity. The exact mechanisms of doxorubicin-induced cardiotoxic side effects remain poorly understood. Even less is known about the impact of doxorubicin treatment on vascular damage. We found that low doses of doxorubicin induced a senescent response in human primary vascular smooth muscle cells (VSMC). We observed that expression of urokinase receptor (uPAR) was upregulated in response to doxorubicin. Furthermore, the level of uPAR expression played a decisive role in developing doxorubicin-induced senescence. uPAR silencing in human VSMC by means of RNA interference as well as uPAR knockout in mouse VSMC resulted in abrogation of doxorubicin-induced cellular senescence. On the contrary, uPAR overexpression promoted VSMC senescence. We further found that proteasomal degradation of telomeric repeat binding factor 2 (TRF2) mediates doxorubicin-induced VSMC senescence. Our results demonstrate that uPAR controls the ubiquitin-proteasome system in VSMC and regulates doxorubicin-induced TRF2 ubiquitination and proteasomal degradation via this mechanism. Therefore, VSMC senescence induced by low doses of doxorubicin may contribute to vascular damage upon doxorubicin treatment. uPAR-mediated TRF2 ubiquitination and proteasomal degradation are further identified as a molecular mechanism underlying this process.
Collapse
Affiliation(s)
- Mahshid Hodjat
- Nephrology Department, Hannover Medical School, Hannover, Germany
| | | | | | | |
Collapse
|
27
|
Martin S, Janouskova H, Dontenwill M. Integrins and p53 pathways in glioblastoma resistance to temozolomide. Front Oncol 2012; 2:157. [PMID: 23120745 PMCID: PMC3484330 DOI: 10.3389/fonc.2012.00157] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 10/16/2012] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor. Surgical resection, postoperative radiotherapy plus concomitant and adjuvant chemotherapy with temozolomide (TMZ) is the standard of care for newly diagnosed glioblastoma. In the past decade, efforts have been made to decipher genomic and core pathway alterations to identify clinically relevant glioblastoma subtypes. Based on these studies and more academic explorations, new potential therapeutic targets were found and several targeting agents were developed. Such molecules should hopefully overcome the resistance of glioblastoma to the current therapy. One of the hallmarks of glioblastoma subtypes was the enrichment of extracellular matrix/invasion-related genes. Integrins, which are cell adhesion molecules important in glioma cell migration/invasion and angiogenesis were one of those genes. Integrins seem to be pertinent therapeutic targets and antagonists recently reached the clinic. Although the p53 pathway appears often altered in glioblastoma, conflicting results can be found in the literature about the clinically relevant impact of the p53 status in the resistance to TMZ. Here, we will summarize the current knowledge on (1) integrin expression, (2) p53 status, and (3) relationship between integrins and p53 to discuss their potential impact on the resistance of glioblastoma to temozolomide.
Collapse
Affiliation(s)
- Sophie Martin
- Laboratory of Biophotonics and Pharmacology, UMR 7213 CNRS, Faculté de Pharmacie, Université de Strasbourg Illkirch, France
| | | | | |
Collapse
|
28
|
Liu J, Huang J, He Y, Liu J, Liao B, Liao G. Genetic variants in the integrin gene predicted microRNA-binding sites were associated with the risk of prostate cancer. Mol Carcinog 2012; 53:280-5. [PMID: 23065910 DOI: 10.1002/mc.21973] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Revised: 09/03/2012] [Accepted: 09/22/2012] [Indexed: 02/05/2023]
Abstract
microRNAs (miRNA) silence target genes through Watson-Crick based binding to the 3'untranslated regions (3'UTR). Thus, polymorphisms in the miRNA-binding sites may disrupt this process and play a potential role in cancer pathogenesis. Integrins have been implicated in the genesis and development of many tumors. This study was designed to evaluate the association between five SNP loci in predicted miRNA-binding sites in five integrin genes and prostate cancer occurrence and prognosis to provide data for screening high-risk Chinese Han individuals. These five polymorphisms were genotyped by using the high-resolution melting method (HRM) in 347 Chinese Han prostate cancer patients with long-time follow-up together with 367 age-matched healthy controls. GC carriers of rs11902171 in ITGAv were associated with a decreased risk of prostate cancer (OR 0.57, 95% CI 0.35-0.93). However, no significant difference was detected in genotype distributions of the five SNP loci in the progression-free survival time of prostate cancer. The ITGAv gene SNP rs11902171 may be potentially associated with the risk of prostate cancer.
Collapse
Affiliation(s)
- Jiaming Liu
- Department of Urology, West China School of Medicine/West China Hospital, Sichuan University, Sichuan Province, P.R., China
| | | | | | | | | | | |
Collapse
|
29
|
Coló GP, Hernández-Varas P, Lock J, Bartolomé RA, Arellano-Sánchez N, Strömblad S, Teixidó J. Focal adhesion disassembly is regulated by a RIAM to MEK-1 pathway. J Cell Sci 2012; 125:5338-52. [PMID: 22946047 DOI: 10.1242/jcs.105270] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cell migration and invasion require regulated turnover of integrin-dependent adhesion complexes. Rap1-GTP-interacting adaptor molecule (RIAM) is an adaptor protein that mediates talin recruitment to the cell membrane, and whose depletion leads to defective melanoma cell migration and invasion. In this study, we investigated the potential involvement of RIAM in focal adhesion (FA) dynamics. RIAM-depleted melanoma and breast carcinoma cells displayed an increased number, size and stability of FAs, which accumulated centrally at the ventral cell surface, a phenotype caused by defective FA disassembly. Impairment in FA disassembly resulting from RIAM knockdown correlated with deficient integrin-dependent mitogen-activated protein kinase kinase (MEK)-Erk1/2 activation and, importantly, overexpression of constitutively active MEK resulted in rescue of FA disassembly and recovery of cell invasion. Furthermore, RIAM-promoted Ras homologue gene family, member A (RhoA) activation following integrin engagement was needed for subsequent Erk1/2 activation. In addition, RhoA overexpression partially rescued the FA phenotype in RIAM-depleted cells, also suggesting a functional role for RhoA downstream of RIAM, but upstream of Erk1/2. RIAM knockdown also led to enhanced phosphorylation of paxillin Tyr118 and Tyr31. However, expression of phosphomimetic and nonphosphorylatable mutants at these paxillin residues indicated that paxillin hyperphosphorylation is a subsequent consequence of the blockade of FA disassembly, but does not cause the FA phenotype. RIAM depletion also weakened the association between FA proteins, suggesting that it has important adaptor roles in the correct assembly of adhesion complexes. Our data suggest that integrin-triggered, RIAM-dependent MEK activation represents a key feedback event required for efficient FA disassembly, which could help explain the role of RIAM in cell migration and invasion.
Collapse
Affiliation(s)
- Georgina P Coló
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), 28040 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
30
|
Smith SD, Enge M, Bao W, Thullberg M, Costa TDF, Olofsson H, Gashi B, Selivanova G, Strömblad S. Protein kinase Cα (PKCα) regulates p53 localization and melanoma cell survival downstream of integrin αv in three-dimensional collagen and in vivo. J Biol Chem 2012; 287:29336-47. [PMID: 22773839 DOI: 10.1074/jbc.m112.341917] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Protein kinase C α (PKCα) is overexpressed in numerous types of cancer. Importantly, PKCα has been linked to metastasis of malignant melanoma in patients. However, it has been unclear how PKCα may be regulated and how it exerts its role in melanoma. Here, we identified a role for PKCα in melanoma cell survival in a three-dimensional collagen model mimicking the in vivo pathophysiology of the dermis. A pathway was identified that involved integrin αv-mediated up-regulation of PKCα and PKCα-dependent regulation of p53 localization, which was connected to melanoma cell survival. Melanoma survival and growth in three-dimensional microenvironments requires the expression of integrin αv, which acts to suppress p53 activity. Interestingly, microarray analysis revealed that PKCα was up-regulated by integrin αv in a three-dimensional microenvironment-dependent manner. Integrin αv was observed to promote a relocalization of endogenous p53 from the nucleus to the cytoplasm upon growth in three-dimensional collagen as well as in vivo, whereas stable knockdown of PKCα inhibited the integrin αv-mediated relocalization of p53. Importantly, knockdown of PKCα also promoted apoptosis in three-dimensional collagen and in vivo, resulting in reduced tumor growth. This indicates that PKCα constitutes a crucial component of the integrin αv-mediated pathway(s) that promote p53 relocalization and melanoma survival.
Collapse
Affiliation(s)
- Stephen D Smith
- Department of Biosciences and Nutrition, Center for Biosciences, Karolinska Institutet, Huddinge SE-141 83, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Genua M, Xu SQ, Buraschi S, Peiper SC, Gomella LG, Belfiore A, Iozzo RV, Morrione A. Proline-rich tyrosine kinase 2 (Pyk2) regulates IGF-I-induced cell motility and invasion of urothelial carcinoma cells. PLoS One 2012; 7:e40148. [PMID: 22859931 PMCID: PMC3408023 DOI: 10.1371/journal.pone.0040148] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 06/01/2012] [Indexed: 12/02/2022] Open
Abstract
The insulin-like growth factor receptor I (IGF-IR) plays an essential role in transformation by promoting cell growth and protecting cancer cells from apoptosis. We have recently demonstrated that the IGF-IR is overexpressed in invasive bladder cancer tissues and promotes motility and invasion of urothelial carcinoma cells. These effects require IGF-I-induced Akt- and MAPK-dependent activation of paxillin. The latter co-localizes with focal adhesion kinases (FAK) at dynamic focal adhesions and is critical for promoting motility of urothelial cancer cells. FAK and its homolog Proline-rich tyrosine kinase 2 (Pyk2) modulate paxillin activation; however, their role in regulating IGF-IR-dependent signaling and motility in bladder cancer has not been established. In this study we demonstrate that FAK was not required for IGF-IR-dependent signaling and motility of invasive urothelial carcinoma cells. On the contrary, Pyk2, which was strongly activated by IGF-I, was critical for IGF-IR-dependent motility and invasion and regulated IGF-I-dependent activation of the Akt and MAPK pathways. Using immunofluorescence and AQUA analysis we further discovered that Pyk2 was overexpressed in bladder cancer tissues as compared to normal tissue controls. Significantly, in urothelial carcinoma tissues there was increased Pyk2 localization in the nuclei as compared to normal tissue controls. These results provide the first evidence of a specific Pyk2 activity in regulating IGF-IR-dependent motility and invasion of bladder cancer cells suggesting that Pyk2 and the IGF-IR may play a critical role in the invasive phenotype in urothelial neoplasia. In addition, Pyk2 and the IGF-IR may serve as novel biomarkers with diagnostic and prognostic significance in bladder cancer.
Collapse
Affiliation(s)
- Marco Genua
- Endocrine Mechanisms and Hormone Action Program, Department of Urology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Shi-Qiong Xu
- Endocrine Mechanisms and Hormone Action Program, Department of Urology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Simone Buraschi
- Cancer Cell Biology and Signaling Program, Department of Pathology, Anatomy and Cell Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Stephen C. Peiper
- Cancer Cell Biology and Signaling Program, Department of Pathology, Anatomy and Cell Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Leonard G. Gomella
- Endocrine Mechanisms and Hormone Action Program, Department of Urology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Antonino Belfiore
- Endocrinology, Department of Health, University of Catanzaro, Catanzaro, Italy
| | - Renato V. Iozzo
- Cancer Cell Biology and Signaling Program, Department of Pathology, Anatomy and Cell Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Andrea Morrione
- Endocrine Mechanisms and Hormone Action Program, Department of Urology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
32
|
Beristain AG, Narala SR, Di Grappa MA, Khokha R. Homotypic RANK signaling differentially regulates proliferation, motility and cell survival in osteosarcoma and mammary epithelial cells. J Cell Sci 2012; 125:943-55. [DOI: 10.1242/jcs.094029] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
RANKL (receptor activator of NF-κB ligand) is a crucial cytokine for regulating diverse biological systems such as innate immunity, bone homeostasis and mammary gland differentiation, operating through activation of its cognate receptor RANK. In these normal physiological processes, RANKL signals through paracrine and/or heterotypic mechanisms where its expression and function is tightly controlled. Numerous pathologies involve RANKL deregulation, such as bone loss, inflammatory diseases and cancer, and aberrant RANK expression has been reported in bone cancer. Here, we investigated the significance of RANK in tumor cells with a particular emphasis on homotypic signaling. We selected RANK-positive mouse osteosarcoma and RANK-negative preosteoblastic MC3T3-E1 cells and subjected them to loss- and gain-of-RANK function analyses. By examining a spectrum of tumorigenic properties, we demonstrate that RANK homotypic signaling has a negligible effect on cell proliferation, but promotes cell motility and anchorage-independent growth of osteosarcoma cells and preosteoblasts. By contrast, establishment of RANK signaling in non-tumorigenic mammary epithelial NMuMG cells promotes their proliferation and anchorage-independent growth, but not motility. Furthermore, RANK activation initiates multiple signaling pathways beyond its canonical target, NF-κB. Among these, biochemical inhibition reveals that Erk1/2 is dominant and crucial for the promotion of anchorage-independent survival and invasion of osteoblastic cells, as well as the proliferation of mammary epithelial cells. Thus, RANK signaling functionally contributes to key tumorigenic properties through a cell-autonomous homotypic mechanism. These data also identify the likely inherent differences between epithelial and mesenchymal cell responsiveness to RANK activation.
Collapse
Affiliation(s)
- Alexander G. Beristain
- Ontario Cancer Institute, Princess Margaret Hospital, 610 University Avenue, Toronto Ontario, M5G 2M9, Canada
| | - Swami R. Narala
- Ontario Cancer Institute, Princess Margaret Hospital, 610 University Avenue, Toronto Ontario, M5G 2M9, Canada
| | - Marco A. Di Grappa
- Ontario Cancer Institute, Princess Margaret Hospital, 610 University Avenue, Toronto Ontario, M5G 2M9, Canada
| | - Rama Khokha
- Ontario Cancer Institute, Princess Margaret Hospital, 610 University Avenue, Toronto Ontario, M5G 2M9, Canada
| |
Collapse
|
33
|
Yu L, Favoino E, Wang Y, Ma Y, Deng X, Wang X. The CSPG4-specific monoclonal antibody enhances and prolongs the effects of the BRAF inhibitor in melanoma cells. Immunol Res 2011; 50:294-302. [PMID: 21717063 DOI: 10.1007/s12026-011-8232-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PLX4032 is a BRAF-selective inhibitor shown to be efficacious in the treatment of melanomas presenting with the BRAF(V600E) mutation. However, favorable responses to treatment are short-lived, and complete remission is rarely observed. Therefore, it is important to identify novel therapies designed to enhance treatment responses and to increase the longevity of initial response to BRAF inhibitors. To this end, we characterized the effects of the 225.28 chondroitin sulfate proteoglycan 4 (CSPG4)-specific monoclonal antibody (mAb) capable of blocking multiple signaling pathways important to cell growth, migration, and survival. Addition of 225.28 to the treatment regimen enhanced the in vitro response magnitude and the duration efficacy of PLX4032 in treating CSPG4(+), BRAF(V600E) melanoma cells (melanoma(BRAF(V600E)/CSPG4+) cells). Data presented in this report demonstrated that (1) treatments comprised of PLX4032 and mAb 225.28 were more effective at inhibiting melanoma(BRAF(V600E)/CSPG4+) cell growth than either agent alone, (2) mAb 225.28 prevented/delayed the development of resistance in melanoma(BRAF(V600E)/CSPG4+) cells to PLX4032, and (3) the mechanism of action of the combination therapy caused a down-regulation in multiple signaling pathways. This study provides a foundation for future investigations designed to improve BRAF inhibitor effectiveness in vitro and in vivo for treating melanoma(BRAF(V600E)/CSPG4+) cells in combination with a CSPG4-specific mAb.
Collapse
Affiliation(s)
- Ling Yu
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | | | | | | | | | | |
Collapse
|
34
|
Integrin-mediated cell-matrix interaction in physiological and pathological blood vessel formation. JOURNAL OF ONCOLOGY 2011; 2012:125278. [PMID: 21941547 PMCID: PMC3175391 DOI: 10.1155/2012/125278] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 07/15/2011] [Indexed: 02/07/2023]
Abstract
Physiological as well as pathological blood vessel formation are fundamentally dependent on cell-matrix interaction. Integrins, a family of major cell adhesion receptors, play a pivotal role in development, maintenance, and remodeling of the vasculature. Cell migration, invasion, and remodeling of the extracellular matrix (ECM) are integrin-regulated processes, and the expression of certain integrins also correlates with tumor progression. Recent advances in the understanding of how integrins are involved in the regulation of blood vessel formation and remodeling during tumor progression are highlighted. The increasing knowledge of integrin function at the molecular level, together with the growing repertoire of integrin inhibitors which allow their selective pharmacological manipulation, makes integrins suited as potential diagnostic markers and therapeutic targets.
Collapse
|
35
|
Abstract
As invading breast carcinoma cells breach their underlying basement membrane, they become confronted with a dense three-dimensional reactive stroma dominated by type I collagen. To develop metastatic capabilities, invading tumor cells must acquire the capacity to negotiate this novel microenvironment. Collagen influences the fate of epithelial cells by inducing apoptosis. However, the mechanisms used by invading tumor cells to evade collagen-induced apoptosis remain to be defined. We demonstrate that membrane type-1 matrix metalloproteinase (MT1-MMP/MMP-14) confers breast cancer cells with the ability to escape apoptosis when embedded in a collagen gel and after orthotopic implantation in vivo. In the absence of MMP-14-dependent proteolysis, type I collagen triggers apoptosis by inducing the expression of the pro-apoptotic Bcl-2-interacting killer in luminal-like breast cancer cells. These findings reveal a new mechanism whereby MMP-14 activity promotes tumor progression by circumventing apoptosis.
Collapse
|
36
|
Bielefeld KA, Amini-Nik S, Whetstone H, Poon R, Youn A, Wang J, Alman BA. Fibronectin and beta-catenin act in a regulatory loop in dermal fibroblasts to modulate cutaneous healing. J Biol Chem 2011; 286:27687-97. [PMID: 21652705 DOI: 10.1074/jbc.m111.261677] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
β-Catenin is an important regulator of dermal fibroblasts during cutaneous wound repair. However, the factors that modulate β-catenin activity in this process are not completely understood. We investigated the role of the extracellular matrix in regulating β-catenin and found an increase in β-catenin-mediated Tcf-dependent transcriptional activity in fibroblasts exposed to various extracellular matrix components. This occurs through an integrin-mediated GSK3β-dependent pathway. The physiologic role of this mechanism was demonstrated during wound repair in extra domain A-fibronectin-deficient mice, which exhibited decreased β-catenin-mediated signaling during the proliferative phase of healing. Extra domain A-fibronectin-deficient mice have wounds that fail at a lower tensile strength and contain fewer fibroblasts compared with wild type mice. This phenotype was rescued by genetic or pharmacologic activation of β-catenin signaling. Because fibronectin is a transcriptional target of β-catenin, this suggests the existence of a feedback loop between these two molecules that regulates dermal fibroblast cell behavior during wound repair.
Collapse
Affiliation(s)
- Kirsten A Bielefeld
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1L7, Canada
| | | | | | | | | | | | | |
Collapse
|
37
|
Hernández-Varas P, Coló GP, Bartolomé RA, Paterson A, Medraño-Fernández I, Arellano-Sánchez N, Cabañas C, Sánchez-Mateos P, Lafuente EM, Boussiotis VA, Strömblad S, Teixidó J. Rap1-GTP-interacting adaptor molecule (RIAM) protein controls invasion and growth of melanoma cells. J Biol Chem 2011; 286:18492-504. [PMID: 21454517 DOI: 10.1074/jbc.m110.189811] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Mig-10/RIAM/lamellipodin (MRL) family member Rap1-GTP-interacting adaptor molecule (RIAM) interacts with active Rap1, a small GTPase that is frequently activated in tumors such as melanoma and prostate cancer. We show here that RIAM is expressed in metastatic human melanoma cells and that both RIAM and Rap1 are required for BLM melanoma cell invasion. RIAM silencing in melanoma cells led to inhibition of tumor growth and to delayed metastasis in a severe combined immunodeficiency xenograft model. Defective invasion of RIAM-silenced melanoma cells arose from impairment in persistent cell migration directionality, which was associated with deficient activation of a Vav2-RhoA-ROCK-myosin light chain pathway. Expression of constitutively active Vav2 and RhoA in cells depleted for RIAM partially rescued their invasion, indicating that Vav2 and RhoA mediate RIAM function. These results suggest that inhibition of cell invasion in RIAM-silenced melanoma cells is likely based on altered cell contractility and cell polarization. Furthermore, we show that RIAM depletion reduces β1 integrin-dependent melanoma cell adhesion, which correlates with decreased activation of both Erk1/2 MAPK and phosphatidylinositol 3-kinase, two central molecules controlling cell growth and cell survival. In addition to causing inhibition of cell proliferation, RIAM silencing led to higher susceptibility to cell apoptosis. Together, these data suggest that defective activation of these kinases in RIAM-silenced cells could account for inhibition of melanoma cell growth and that RIAM might contribute to the dissemination of melanoma cells.
Collapse
Affiliation(s)
- Pablo Hernández-Varas
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas, 28040 Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Berchtold D, Fesser S, Bachmann G, Kaiser A, Eilert JC, Frohns F, Sadoni N, Muck J, Kremmer E, Eick D, Layer PG, Zink D. Nuclei of chicken neurons in tissues and three-dimensional cell cultures are organized into distinct radial zones. Chromosome Res 2011; 19:165-82. [PMID: 21249442 DOI: 10.1007/s10577-010-9182-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 12/22/2010] [Accepted: 12/22/2010] [Indexed: 12/15/2022]
Abstract
We used chicken retinospheroids (RS) to study the nuclear architecture of vertebrate cells in a three-dimensional (3D) cell culture system. The results showed that the different neuronal cell types of RS displayed an extreme form of radial nuclear organization. Chromatin was arranged into distinct radial zones which became already visible after DAPI staining. The distinct zones were enriched in different chromatin modifications and in different types of chromosomes. Active isoforms of RNA polymerase II were depleted in the outermost zone. Also chromocenters and nucleoli were radially aligned in the nuclear interior. The splicing factor SC35 was enriched at the central zone and did not show the typical speckled pattern of distribution. Evaluation of neuronal and non-neuronal chicken tissues showed that the highly ordered form of radial nuclear organization was also present in neuronal chicken tissues. Furthermore, the data revealed that the neuron-specific nuclear organization was remodeled when cells spread on a flat substrate. Monolayer cultures of a chicken cell line did not show this extreme form of radial organization. Rather, such monolayer cultures displayed features of nuclear organization which have been described before for many different types of monolayer cells. The finding that an extreme form radial nuclear organization, which has not been described before, is present in RS and tissues, but not in cells spread on a flat substrate, suggests that it would be important to complement studies on nuclear architecture performed with monolayer cells by studies on 3D cell culture systems and tissues.
Collapse
Affiliation(s)
- Doris Berchtold
- Department Biologie II, Ludwig-Maximilians-Universität (LMU) München, Biozentrum, Grosshadernerstr. 2, 82152, Planegg-Martinsried, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Li Z, Zhang H, Lundin L, Thullberg M, Liu Y, Wang Y, Claesson-Welsh L, Strömblad S. p21-activated kinase 4 phosphorylation of integrin beta5 Ser-759 and Ser-762 regulates cell migration. J Biol Chem 2010; 285:23699-710. [PMID: 20507994 DOI: 10.1074/jbc.m110.123497] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Modulation of integrin alphavbeta5 regulates vascular permeability, angiogenesis, and tumor dissemination. In addition, we previously found a role for p21-activated kinase 4 (PAK4) in selective regulation of integrin alphavbeta5-mediated cell motility (Zhang, H., Li, Z., Viklund, E. K., and Strömblad, S. (2002) J. Cell Biol. 158, 1287-1297). This report focuses on the molecular mechanisms of this regulation. We here identified a unique PAK4-binding membrane-proximal integrin beta5-SERS-motif involved in controlling cell attachment and migration. We also mapped the integrin beta5-binding site within PAK4. We found that PAK4 binding to integrin beta5 was not sufficient to promote cell migration, but that PAK4 kinase activity was required for PAK4 promotion of cell motility. Importantly, PAK4 specifically phosphorylated the integrin beta5 subunit at Ser-759 and Ser-762 within the beta5-SERS-motif. Point mutation of these two serine residues abolished the PAK4-induced cell migration, indicating a functional role for these phosphorylations in migration. Our results may give important leads to the functional regulation of integrin alphavbeta5, with implications for vascular permeability, angiogenesis, and cancer dissemination.
Collapse
Affiliation(s)
- Zhilun Li
- Center for Biosciences, Department of Biosciences and Nutrition, Karolinska Institutet, 141 83 Huddinge, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Metalli D, Lovat F, Tripodi F, Genua M, Xu SQ, Spinelli M, Alberghina L, Vanoni M, Baffa R, Gomella LG, Iozzo RV, Morrione A. The insulin-like growth factor receptor I promotes motility and invasion of bladder cancer cells through Akt- and mitogen-activated protein kinase-dependent activation of paxillin. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2997-3006. [PMID: 20395438 DOI: 10.2353/ajpath.2010.090904] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The insulin-like growth factor receptor I (IGF-IR) plays an essential role in transformation by promoting cell growth and protecting cancer cells from apoptosis. Aberrant IGF-IR signaling is implicated in several types of tumors, including carcinomas of the lung, breast, prostate, pancreas, liver, and colon. However, the contribution of the IGF-IR to the development of the transformed phenotype in urothelial cells has not been clearly established. In this study we demonstrated that the IGF-IR is overexpressed in invasive bladder cancer tissues compared with nonmalignant controls. We have investigated the role of the IGF-IR in bladder cancer by using urothelial carcinoma-derived 5637 and T24 cells. Although activation of the IGF-IR did not appreciably affect their growth, it did promote migration and stimulate in vitro wound closure and invasion. These effects required the activation of the Akt and Mitogen-activated protein kinase (MAPK) pathways as well as IGF-I-induced Akt- and MAPK-dependent phosphorylation of paxillin, which relocated at dynamic focal adhesions and was necessary for promoting motility in bladder cancer cells. Our results provide the first evidence for a role of the IGF-IR in motility and invasion of bladder cancer cells and support the hypothesis that the IGF-IR may play a critical role in the establishment of the invasive phenotype in urothelial neoplasia. Thus, the IGF-IR may also serve as a novel biomarker for bladder cancer.
Collapse
Affiliation(s)
- David Metalli
- Department of Urology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
The integrin family of cell adhesion receptors regulates a diverse array of cellular functions crucial to the initiation, progression and metastasis of solid tumours. The importance of integrins in several cell types that affect tumour progression has made them an appealing target for cancer therapy. Integrin antagonists, including the alphavbeta3 and alphavbeta5 inhibitor cilengitide, have shown encouraging activity in Phase II clinical trials and cilengitide is currently being tested in a Phase III trial in patients with glioblastoma. These exciting clinical developments emphasize the need to identify how integrin antagonists influence the tumour and its microenvironment.
Collapse
Affiliation(s)
- Jay S Desgrosellier
- Department of Pathology, Moores University of California at San Diego Cancer Center, La Jolla, 92093-0803, United States
| | | |
Collapse
|
42
|
Desgrosellier JS, Barnes LA, Shields DJ, Huang M, Lau SK, Prévost N, Tarin D, Shattil SJ, Cheresh DA. An integrin alpha(v)beta(3)-c-Src oncogenic unit promotes anchorage-independence and tumor progression. Nat Med 2009; 15:1163-9. [PMID: 19734908 PMCID: PMC2759406 DOI: 10.1038/nm.2009] [Citation(s) in RCA: 235] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Accepted: 06/19/2009] [Indexed: 01/13/2023]
Abstract
Integrins regulate adhesion-dependent growth, survival and invasion of tumor cells. In particular, expression of integrin alpha(v)beta(3) is associated with progression of a variety of human tumors. Here we reveal a previously undescribed adhesion-independent role for integrin alpha(v)beta(3) in pancreatic cancer and other carcinomas. Specifically, alpha(v)beta(3) expressed in carcinoma cells enhanced anchorage-independent tumor growth in vitro and increased lymph node metastases in vivo. These effects required recruitment of c-Src to the beta(3) integrin cytoplasmic tail, leading to c-Src activation, Crk-associated substrate (CAS) phosphorylation and tumor cell survival that, unexpectedly, was independent of cell adhesion or focal adhesion kinase (FAK) activation. Pharmacological blockade of c-Src kinase activity or decreased expression of endogenous alpha(v)beta(3) integrin or c-Src not only inhibited anchorage-independent growth but also suppressed metastasis in vivo, yet these manipulations did not affect tumor cell migration or invasion. These data define an unexpected role for an integrin as a mediator of anchorage independence, suggesting that an alpha(v)beta(3)-c-Src signaling module may account for the aggressive behavior of integrin alpha(v)beta(3)-expressing tumors in humans.
Collapse
Affiliation(s)
- Jay S Desgrosellier
- Department of Pathology, Moores UCSD Cancer Center University of California, San Diego, La Jolla, CA 92093, USA
| | - Leo A Barnes
- Department of Pathology, Moores UCSD Cancer Center University of California, San Diego, La Jolla, CA 92093, USA
| | - David J Shields
- Department of Pathology, Moores UCSD Cancer Center University of California, San Diego, La Jolla, CA 92093, USA
| | - Miller Huang
- Department of Pathology, Moores UCSD Cancer Center University of California, San Diego, La Jolla, CA 92093, USA
| | - Steven K Lau
- Department of Pathology, Moores UCSD Cancer Center University of California, San Diego, La Jolla, CA 92093, USA
| | - Nicolas Prévost
- Division of Hematology-Oncology, Department of Medicine University of California, San Diego, La Jolla, CA 92093, USA
| | - David Tarin
- Department of Pathology, Moores UCSD Cancer Center University of California, San Diego, La Jolla, CA 92093, USA
| | - Sanford J Shattil
- Division of Hematology-Oncology, Department of Medicine University of California, San Diego, La Jolla, CA 92093, USA
| | - David A Cheresh
- Department of Pathology, Moores UCSD Cancer Center University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
43
|
SPARC in cancer biology: its role in cancer progression and potential for therapy. Drug Resist Updat 2008; 11:231-46. [PMID: 18849185 DOI: 10.1016/j.drup.2008.08.005] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 08/25/2008] [Accepted: 08/26/2008] [Indexed: 02/06/2023]
Abstract
The ability to effectively target a tumor to achieve complete regression and cure is the ultimate goal that drives our need to better understand tumor biology. Recently, SPARC has generated considerable interest as a multi-faceted protein that belongs to a family of matricellular proteins. It functions not only to modulate cell-cell and cell-matrix interactions, but its de-adhesive and growth inhibitory properties in non-transformed cells have led to studies to assess its role in cancer. Its divergent actions reflect the complexity of this protein, because in certain types of cancers, such as melanomas and gliomas, SPARC is associated with a highly aggressive tumor phenotype, while in others, mainly ovarian, neuroblastomas and colorectal cancers, SPARC may function as a tumor suppressor. Recent studies have also demonstrated a role for SPARC in sensitizing therapy-resistant cancers. Here, the role of SPARC in cancer progression and its potential application in cancer therapy is discussed.
Collapse
|
44
|
Zhang Y, Shu L, Chen X. Syntaxin 6, a regulator of the protein trafficking machinery and a target of the p53 family, is required for cell adhesion and survival. J Biol Chem 2008; 283:30689-98. [PMID: 18779328 DOI: 10.1074/jbc.m801711200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The p53 family consists of p53, p63, and p73. It has been well characterized that all of the p53 family proteins are transcription factors and capable of regulating cell cycle and apoptosis. To determine whether the p53 family exerts tumor suppression by other mechanisms, we set to identify novel p53 family target genes. Here, we found that the gene encoding STX6 (syntaxin 6), a vesicle transporter protein, is directly regulated by each of the p53 family proteins. In addition, STX6 can be induced by DNA damage and Mdm2 inhibitor Nutlin-3 in a p53-dependent manner. To examine how STX6 mediates the activity of the p53 family, STX6 is inducibly overexpressed or knocked down in various cell lines. We found that overexpression of STX6 alone has limited effect on cell proliferation. In contrast, we found that knockdown of STX6 inhibits cell proliferation and survival. We also found that knockdown of STX6 leads to cell cycle arrest and apoptosis. Interestingly, we found that p53 is necessary for STX6 knockdown-induced cell cycle arrest and apoptosis. Furthermore, we found that STX6 is necessary for proper expression of focal adhesion kinase and integrin alpha5 adhesion receptor. Consistent with this observation, STX6 knockdown inhibits cell adhesion. Together, we postulate that STX6 is an effector and a modulator of the p53 family in the regulation of cell adhesion and survival.
Collapse
Affiliation(s)
- Yanhong Zhang
- Center for Comparative Oncology, University of California, Davis, California 95616, USA
| | | | | |
Collapse
|
45
|
Hirai Y, Nelson CM, Yamazaki K, Takebe K, Przybylo J, Madden B, Radisky DC. Non-classical export of epimorphin and its adhesion to alphav-integrin in regulation of epithelial morphogenesis. J Cell Sci 2007; 120:2032-43. [PMID: 17535848 DOI: 10.1242/jcs.006247] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Epimorphin (also known as syntaxin 2) acts as an epithelial morphogen when secreted by stromal cells of the mammary gland, lung, liver, colon, pancreas and other tissues, but the same molecule functions within the cell to mediate membrane fusion. How this molecule, which lacks a signal sequence and contains a transmembrane domain at the C-terminus, translocates across the plasma membrane and is secreted to become a morphogen, and how it initiates morphogenic events is not clear. Here, we show that epimorphin is secreted through a non-classical mechanism, similar to that previously described for secretion of the leaderless protein FGF1, and we identify the key molecular elements responsible for translocation and secretion from the cell. We also show that secreted epimorphin binds to alphav-integrin-containing receptors on target epithelial cells, leading to activation of specific downstream signaling pathways and induction of epithelial morphogenesis. These findings provide key insight into how epimorphin functions as an epithelial morphogen.
Collapse
Affiliation(s)
- Yohei Hirai
- Department of Morphoregulation, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| | | | | | | | | | | | | |
Collapse
|
46
|
Said N, Najwer I, Motamed K. Secreted protein acidic and rich in cysteine (SPARC) inhibits integrin-mediated adhesion and growth factor-dependent survival signaling in ovarian cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:1054-63. [PMID: 17322388 PMCID: PMC1864881 DOI: 10.2353/ajpath.2007.060903] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The matricellular glycoprotein SPARC (secreted protein acidic and rich in cysteine) has been accorded major roles in regulation of cell adhesion and proliferation, as well as tumorigenesis and metastasis. We have recently reported that in addition to its potent antiproliferative and proapoptotic functions, SPARC also abrogates ovarian carcinoma cell adhesion, a key step in peritoneal implantation. However, the underlying molecular mechanism through which SPARC ameliorates peritoneal ovarian carcinomatosis seems to be multifaceted and has yet to be delineated. Herein, we show that SPARC significantly inhibited integrin-mediated ovarian cancer cell adhesion to extracellular matrix proteins, as well as to peritoneal mesothelial cells. This counteradhesive effect of SPARC was shown to be mediated in part through significant attenuation of cell surface expression and clustering of alpha(v)-integrin subunit, alpha(v)beta(3)- and alpha(v)beta(5)-heterodimers, and beta(1)-subunit, albeit to a lesser extent, in ovarian cancer cells. Moreover, SPARC significantly suppressed both anchorage-dependent and -independent activation of AKT and mitogen-acti-vated protein kinase survival signaling pathways in ovarian cancer cells in response to serum and epidermal growth factor stimulation. In summary, we have identified a novel role of SPARC as a negative regulator of both integrin-mediated adhesion and growth factor-stimulated survival signaling pathways in ovarian cancer.
Collapse
Affiliation(s)
- Neveen Said
- Vascular Biology Center, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | |
Collapse
|
47
|
Smalley KSM, Lioni M, Herlyn M. Life isn't flat: taking cancer biology to the next dimension. In Vitro Cell Dev Biol Anim 2007; 42:242-7. [PMID: 17163781 DOI: 10.1290/0604027.1] [Citation(s) in RCA: 219] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Accepted: 07/11/2006] [Indexed: 12/29/2022]
Abstract
Classically, most cell culture experiments have been performed under adherent 2D conditions. Cells in the human body grow within an organized 3D matrix, surrounded by other cells. The behavior of individual cells is controlled through their interactions with their immediate neighbors and the extracellular matrix. The complex summation of these multiple signals determines whether a given cell undergoes differentiation, apoptosis, proliferation, or invasion. In 2D culture many of these complex interactions are lost. As a result, there are a growing number of studies which report differences in phenotype, cellular signaling, cell migration, and drug responses when the same cells are grown under 2D or 3D culture conditions. One potential application of these techniques is to anticancer drug discovery, which has long been hampered by the lack of good preclinical models. Compounds with good antitumor activity in 2D cell culture models often fail to translate into the clinic. Here we suggest that the response of cancer cells to drugs is determined in part by the 3D tumor microenvironment and discuss models to re-create the 3D tumor microenvironment in vitro. It is likely that the adoption of these and other 3D models will allow us to more closely re-create the behavior of the tumor in vivo which may lead to identifying better anticancer drug candidates at an earlier stage of development.
Collapse
|
48
|
Toriseva MJ, Ala-aho R, Karvinen J, Baker AH, Marjomäki VS, Heino J, Kähäri VM. Collagenase-3 (MMP-13) Enhances Remodeling of Three-Dimensional Collagen and Promotes Survival of Human Skin Fibroblasts. J Invest Dermatol 2007; 127:49-59. [PMID: 16917496 DOI: 10.1038/sj.jid.5700500] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Collagenase-3 (MMP-13) is a matrix metalloproteinase capable of cleaving a multitude of extracellular matrix proteins in addition to fibrillar collagens. Human MMP-13 is expressed by fibroblasts in chronic cutaneous ulcers, but not in normally healing adult skin wounds. However, MMP-13 is produced by fibroblasts in adult gingival and in fetal skin wounds characterized by rapid collagen remodeling and scarless healing. Here, we have examined the role of human MMP-13 in remodeling of three-dimensional (3D) collagenous matrix by primary adult human skin fibroblasts. The high level of human MMP-13 expression by fibroblasts achieved by adenoviral gene delivery resulted in potent enhancement of remodeling and contraction of 3D collagen. Fibroblasts expressing MMP-13 in 3D collagen possessed altered filamentous actin morphology with patch-like actin distribution in cell extensions. The expression of MMP-13 promotes survival and proliferation of fibroblasts in floating collagen gel, and results in activation of Akt and extracellular signal-regulated kinase-1/2 by these cells. The results provide evidence for a novel role for human MMP-13 in regulating dermal fibroblast survival, proliferation, and interaction in 3D collagen, which may be an important survival mechanism for fibroblasts in chronic skin ulcers and contribute to scarless healing of adult gingival and fetal skin wounds.
Collapse
|
49
|
Besch R, Berking C, Kammerbauer C, Degitz K. Inhibition of urokinase-type plasminogen activator receptor induces apoptosis in melanoma cells by activation of p53. Cell Death Differ 2006; 14:818-29. [PMID: 17110957 DOI: 10.1038/sj.cdd.4402065] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The urokinase-type plasminogen activator receptor (uPAR) is involved in several biological processes, including proteolysis, adhesion, migration and inflammation. Increased expression of uPAR is associated with metastasis in several tumor types. We studied the biological role of uPAR in melanoma and found that inhibition of uPAR via RNA interference induced massive death in three different metastatic cell lines. Annexin-V staining and caspase activation analysis revealed induction of the mitochondrial apoptotic pathway. The expression of members of the Bcl-2 family (Bax, Bcl-2, Bak and Bcl-x(L)) was changed in a pro-apoptotic manner. uPAR inhibition induced the expression of the tumor suppressor p53 and of its downstream target gene p21. Inhibition of p53 rescued cells from apoptosis indicating that p53 was critical for apoptosis induction. Apoptosis was observed in melanoma cells carrying activating BRAF mutations and occurred in the presence of extracellular signal-regulated kinase (ERK) phosphorylation. uPAR can activate focal adhesion kinase (FAK), which is implicated in adhesion-dependent tumor cell survival. However, inhibition of FAK did not induce apoptosis. Our data suggest a new function of uPAR acting as a survival factor for melanoma by downregulating p53. Inhibition of uPAR induces a pro-apoptotic signalling pathway via p53 that is independent of ERK or FAK signalling. These findings may offer new treatment strategies for metastatic melanoma.
Collapse
Affiliation(s)
- R Besch
- Department of Dermatology and Allergology, Ludwig-Maximilian University, Munich, Germany.
| | | | | | | |
Collapse
|
50
|
Monami G, Gonzalez EM, Hellman M, Gomella LG, Baffa R, Iozzo RV, Morrione A. Proepithelin promotes migration and invasion of 5637 bladder cancer cells through the activation of ERK1/2 and the formation of a paxillin/FAK/ERK complex. Cancer Res 2006; 66:7103-10. [PMID: 16849556 DOI: 10.1158/0008-5472.can-06-0633] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The growth factor proepithelin (also known as progranulin, acrogranin, PC-derived growth factor, or granulin-epithelin precursor) is a secreted glycoprotein that functions as an important regulator of cell growth, migration, and transformation. Proepithelin is overexpressed in a great variety of cancer cell lines and clinical specimens of breast, ovarian, and renal cancer as well as glioblastomas. In this study, we have investigated the effects of proepithelin on bladder cancer cells using human recombinant proepithelin purified to homogeneity from 293-EBNA cells. Although proepithelin did not appreciably affect cell growth, it did promote migration of 5637 bladder cancer cells and stimulate in vitro wound closure and invasion. These effects required the activation of the mitogen-activated protein kinase pathway and paxillin, which upon proepithelin stimulation formed a complex with focal adhesion kinase and active extracellular signal-regulated kinase. Our results provide the first evidence for a role of proepithelin in stimulating migration and invasion of bladder cancer cells, and support the hypothesis that this growth factor may play a critical role in the establishment of the invasive phenotype.
Collapse
Affiliation(s)
- Giada Monami
- Department of Urology, Anatomy and Cell Biology and Cellular Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | |
Collapse
|