1
|
Hoffmann H, Wartenberg M, Vorlova S, Karl-Schöller F, Kallius M, Reinhardt O, Öztürk A, Schuhmair LS, Burkhardt V, Gätzner S, Scheld D, Nandigama R, Zernecke A, Herterich S, Ergün S, Rosenwald A, Henke E. Normalization of Snai1-mediated vessel dysfunction increases drug response in cancer. Oncogene 2024; 43:2661-2676. [PMID: 39095583 PMCID: PMC11347376 DOI: 10.1038/s41388-024-03113-1] [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: 12/14/2023] [Revised: 06/10/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Blood vessels in tumors are often dysfunctional. This impairs the delivery of therapeutic agents to and distribution among the cancer cells. Subsequently, treatment efficacy is reduced, and dose escalation can increase adverse effects on non-malignant tissues. The dysfunctional vessel phenotypes are attributed to aberrant pro-angiogenic signaling, and anti-angiogenic agents can ameliorate traits of vessel dysfunctionality. However, they simultaneously reduce vessel density and thereby impede drug delivery and distribution. Exploring possibilities to improve vessel functionality without compromising vessel density in the tumor microenvironment, we evaluated transcription factors (TFs) involved in epithelial-mesenchymal transition (EMT) as potential targets. Based on similarities between EMT and angiogenic activation of endothelial cells, we hypothesized that these TFs, Snai1 in particular, might serve as key regulators of vessel dysfunctionality. In vitro, experiments demonstrated that Snai1 (similarly Slug and Twist1) regulates endothelial permeability, permissiveness for tumor cell transmigration, and tip/stalk cell formation. Endothelial-specific, heterozygous knock-down of Snai1 in mice improved vascular quality in implanted tumors. This resulted in better oxygenation and reduced metastasis. Notably, the tumors in Snai1KD mice responded significantly better to chemotherapeutics as drugs were transported into the tumors at strongly increased rates and more homogeneously distributed. Thus, we demonstrate that restoring vessel homeostasis without affecting vessel density is feasible in malignant tumors. Combining such vessel re-engineering with anti-cancer drugs allows for strategic treatment approaches that reduce treatment toxicity on non-malignant tissues.
Collapse
Affiliation(s)
- Helene Hoffmann
- Institute of Anatomy and Cell Biology, Universität Würzburg, Koellikerstrasse 6, 97070, Würzburg, Germany
- Graduate School of Life Science, Universität Würzburg, Josef-Schneider-Strasse 2, 97082, Würzburg, Germany
| | - Martin Wartenberg
- Institute of Pathology, Universität Würzburg, and Comprehensive Cancer Center Mainfranken (CCCMF), Josef-Schneider-Strasse 2, 97082, Würzburg, Germany
- Institute of Tissue Medicine and Pathology (ITMP), Universität Bern, Murtenstrasse 31, 3008, Bern, Switzerland
| | - Sandra Vorlova
- Institute of Experimental Biomedicine II, Universitätsklinikum Würzburg, Josef-Schneider-Strasse 2/D16, 97082, Würzburg, Germany
| | - Franziska Karl-Schöller
- Institute of Anatomy and Cell Biology, Universität Würzburg, Koellikerstrasse 6, 97070, Würzburg, Germany
| | - Matthias Kallius
- Institute of Anatomy and Cell Biology, Universität Würzburg, Koellikerstrasse 6, 97070, Würzburg, Germany
- Graduate School of Life Science, Universität Würzburg, Josef-Schneider-Strasse 2, 97082, Würzburg, Germany
| | - Oliver Reinhardt
- Institute of Anatomy and Cell Biology, Universität Würzburg, Koellikerstrasse 6, 97070, Würzburg, Germany
| | - Asli Öztürk
- Institute of Anatomy and Cell Biology, Universität Würzburg, Koellikerstrasse 6, 97070, Würzburg, Germany
| | - Leah S Schuhmair
- Institute of Anatomy and Cell Biology, Universität Würzburg, Koellikerstrasse 6, 97070, Würzburg, Germany
| | - Verena Burkhardt
- Institute of Anatomy and Cell Biology, Universität Würzburg, Koellikerstrasse 6, 97070, Würzburg, Germany
| | - Sabine Gätzner
- Chair Tissue Engineering and Regenerative Medicine (TERM), Universitätsklinikum Würzburg, Roentgenring 11, 97070, Würzburg, Germany
| | - Daniela Scheld
- Zentrallabor, Universitätsklinikum Würzburg, Josef-Schneider-Strasse 2, 97082, Würzburg, Germany
| | - Rajender Nandigama
- Institute of Anatomy and Cell Biology, Universität Würzburg, Koellikerstrasse 6, 97070, Würzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine II, Universitätsklinikum Würzburg, Josef-Schneider-Strasse 2/D16, 97082, Würzburg, Germany
| | - Sabine Herterich
- Zentrallabor, Universitätsklinikum Würzburg, Josef-Schneider-Strasse 2, 97082, Würzburg, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Universität Würzburg, Koellikerstrasse 6, 97070, Würzburg, Germany
| | - Andreas Rosenwald
- Institute of Pathology, Universität Würzburg, and Comprehensive Cancer Center Mainfranken (CCCMF), Josef-Schneider-Strasse 2, 97082, Würzburg, Germany
| | - Erik Henke
- Institute of Anatomy and Cell Biology, Universität Würzburg, Koellikerstrasse 6, 97070, Würzburg, Germany.
- Graduate School of Life Science, Universität Würzburg, Josef-Schneider-Strasse 2, 97082, Würzburg, Germany.
| |
Collapse
|
2
|
Wang L, Sheth V, Liu K, Panja P, Frickenstein AN, He Y, Yang W, Thomas AG, Jamei MH, Park J, Lyu S, Donahue ND, Chen WR, Bhattacharya R, Mukherjee P, Wilhelm S. Primary Human Breast Cancer-Associated Endothelial Cells Favor Interactions with Nanomedicines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403986. [PMID: 38663008 PMCID: PMC11239290 DOI: 10.1002/adma.202403986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/19/2024] [Indexed: 05/04/2024]
Abstract
Cancer nanomedicines predominately rely on transport processes controlled by tumor-associated endothelial cells to deliver therapeutic and diagnostic payloads into solid tumors. While the dominant role of this class of endothelial cells for nanoparticle transport and tumor delivery is established in animal models, the translational potential in human cells needs exploration. Using primary human breast cancer as a model, the differential interactions of normal and tumor-associated endothelial cells with clinically relevant nanomedicine formulations are explored and quantified. Primary human breast cancer-associated endothelial cells exhibit up to ≈2 times higher nanoparticle uptake than normal human mammary microvascular endothelial cells. Super-resolution imaging studies reveal a significantly higher intracellular vesicle number for tumor-associated endothelial cells, indicating a substantial increase in cellular transport activities. RNA sequencing and gene expression analysis indicate the upregulation of transport-related genes, especially motor protein genes, in tumor-associated endothelial cells. Collectively, the results demonstrate that primary human breast cancer-associated endothelial cells exhibit enhanced interactions with nanomedicines, suggesting a potentially significant role for these cells in nanoparticle tumor delivery in human patients. Engineering nanoparticles that leverage the translational potential of tumor-associated endothelial cell-mediated transport into human solid tumors may lead to the development of safer and more effective clinical cancer nanomedicines.
Collapse
Affiliation(s)
- Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Vinit Sheth
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Prasanta Panja
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Alex N Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Yuxin He
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Wen Yang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Abigail G Thomas
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Mohammad Hasan Jamei
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Jeesoo Park
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Shanxin Lyu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Nathan D Donahue
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Institute for Biomedical Engineering, Science and Technology (IBEST), Norman, OK, 73019, USA
| |
Collapse
|
3
|
Elmoselhi AB, Seif Allah M, Bouzid A, Ibrahim Z, Venkatachalam T, Siddiqui R, Khan NA, Hamoudi RA. Circulating microRNAs as potential biomarkers of early vascular damage in vitamin D deficiency, obese, and diabetic patients. PLoS One 2023; 18:e0283608. [PMID: 36952563 PMCID: PMC10035929 DOI: 10.1371/journal.pone.0283608] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 03/13/2023] [Indexed: 03/25/2023] Open
Abstract
Vitamin D3 deficiency, obesity, and diabetes mellitus (DM) have been shown to increase the risk of cardiovascular diseases (CVDs). However, the early detection of vascular damage in those patients is still difficult to ascertain. MicroRNAs (miRNAs) are recognized to play a critical role in initiation and pathogenesis of vascular dysfunction. Herein, we aimed to identify circulating miRNA biomarkers of vascular dysfunction as early predictors of CVDs. We have recruited 23 middle-aged Emiratis patients with the following criteria: A healthy control group with vitamin D ≥ 20ng, and BMI < 30 (C1 group = 11 individuals); A vitamin D deficiency (Vit D level ≤ 20 ng) and obese (BMI ≥ 30) group (A1 group = 9 patients); A vitamin D deficiency, obese, plus DM (A2 group = 3 patients). Arterial stiffness via pulse wave velocity (PWV) was measured and the whole transcriptome analysis with qPCR validation for miRNA in plasma samples were tested. PWV relative to age was significantly higher in A1 group 19.4 ± 4.7 m/s and A2 group 18.3 ± 1.3 m/s compared to controls 14.7 ± 2.1 m/s (p < 0.05). Similar patterns were also observed in the Augmentation pressure (AP) and Alx%. Whole RNA-Sequencing revealed miR-182-5p; miR-199a-5p; miR-193a-5p; and miR-155-5p were differentially over-expressed (logFC > 1.5) in high-risk patients for CVDs vs healthy controls. Collectively, our result indicates that four specific circulating miRNA signature, may be utilized as non-invasive, diagnostic and prognostic biomarkers for early vascular damage in patients suffering from vitamin D deficiency, obesity and DM.
Collapse
Affiliation(s)
- Adel B. Elmoselhi
- College of Medicine, University of Sharjah, Sharjah, UAE
- Sharjah Institute of Medical Research, College of Medicine, University of Sharjah, Sharjah, UAE
| | - Mohamed Seif Allah
- College of Medicine, University of Sharjah, Sharjah, UAE
- Cardiology Department, University Hospital Sharjah, Sharjah, UAE
| | - Amal Bouzid
- Sharjah Institute of Medical Research, College of Medicine, University of Sharjah, Sharjah, UAE
| | - Zeinab Ibrahim
- Sharjah Institute of Medical Research, College of Medicine, University of Sharjah, Sharjah, UAE
| | - Thenmozhi Venkatachalam
- Sharjah Institute of Medical Research, College of Medicine, University of Sharjah, Sharjah, UAE
| | - Ruqaiyyah Siddiqui
- College of Arts and Sciences, American University of Sharjah, University City, Sharjah, UAE
| | - Naveed Ahmed Khan
- College of Medicine, University of Sharjah, Sharjah, UAE
- Sharjah Institute of Medical Research, College of Medicine, University of Sharjah, Sharjah, UAE
| | - Rifat A. Hamoudi
- College of Medicine, University of Sharjah, Sharjah, UAE
- Sharjah Institute of Medical Research, College of Medicine, University of Sharjah, Sharjah, UAE
| |
Collapse
|
4
|
Differential angiogenesis of bone and muscle endothelium in aging and inflammatory processes. Commun Biol 2023; 6:126. [PMID: 36721025 PMCID: PMC9889796 DOI: 10.1038/s42003-023-04515-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 01/20/2023] [Indexed: 02/01/2023] Open
Abstract
Different tissues have different endothelial features, however, the implications of this heterogeneity in pathological responses are not clear yet. "Inflamm-aging" has been hypothesized as a possible trigger of diseases, including osteoarthritis (OA) and sarcopenia, often present in the same patient. To highlight a possible contribution of organ-specific endothelial cells (ECs), we compare ECs derived from bone and skeletal muscle of the same OA patients. OA bone ECs show a pro-inflammatory signature and higher angiogenic sprouting as compared to muscle ECs, in control conditions and stimulated with TNFα. Furthermore, growth of muscle but not bone ECs decreases with increasing patient age and systemic inflammation. Overall, our data demonstrate that inflammatory conditions in OA patients differently affect bone and muscle ECs, suggesting that inflammatory processes increase angiogenesis in subchondral bone while associated systemic low-grade inflammation impairs angiogenesis in muscle, possibly highlighting a vascular trigger linking OA and sarcopenia.
Collapse
|
5
|
Catulin reporter marks a heterogeneous population of invasive breast cancer cells with some demonstrating plasticity and participating in vascular mimicry. Sci Rep 2022; 12:12673. [PMID: 35879327 PMCID: PMC9314412 DOI: 10.1038/s41598-022-16802-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 07/15/2022] [Indexed: 11/30/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer in women worldwide. The activation of partial or more complete epithelial–mesenchymal transition in cancer cells enhances acquisition of invasive behaviors and expands their generation of cancer stem cells. Increased by EMT plasticity of tumor cells could promote vascular mimicry, a newly defined pattern of tumor microvascularization by which aggressive tumor cells can form vessel-like structures themselves. VM is strongly associated with a poor prognosis, but biological features of tumor cells that form VM remains unknown. Here we show that catulin is expressed in human BC samples and its expression correlates with the tumor progression. Ablation of catulin in hBC cell lines decreases their invasive potential in the 3D assays. Using a novel catulin promoter based reporter we tracked and characterized the small population of invasive BC cells in xenograft model. RNAseq analysis revealed enrichment in genes important for cellular movement, invasion and interestingly for tumor-vasculature interactions. Analysis of tumors unveiled that catulin reporter marks not only invasive cancer cells but also rare population of plastic, MCAM positive cancer cells that participate in vascular mimicry. Ablation of catulin in the xenograft model revealed deregulation of genes involved in cellular movement, and adhesive properties with striking decrease in CD44 which may impact stemness potential, and plasticity of breast cancer cells. These findings show directly that some plastic tumor cells can change the fate into endothelial-like, expressing MCAM and emphasize the importance of catulin in this process and breast cancer progression.
Collapse
|
6
|
Kwon NY, Sung SH, Sung HK, Park JK. Anticancer Activity of Bee Venom Components against Breast Cancer. Toxins (Basel) 2022; 14:toxins14070460. [PMID: 35878198 PMCID: PMC9318616 DOI: 10.3390/toxins14070460] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/27/2022] [Accepted: 07/02/2022] [Indexed: 12/10/2022] Open
Abstract
While the survival rate has increased due to treatments for breast cancer, the quality of life has decreased because of the side effects of chemotherapy. Various toxins are being developed as alternative breast cancer treatments, and bee venom is drawing attention as one of them. We analyzed the effect of bee venom and its components on breast cancer cells and reviewed the mechanism underlying the anticancer effects of bee venom. Data up to March 2022 were searched from PubMed, EMBASE, OASIS, KISS, and Science Direct online databases, and studies that met the inclusion criteria were reviewed. Among 612 studies, 11 were selected for this research. Diverse drugs were administered, including crude bee venom, melittin, phospholipase A2, and their complexes. All drugs reduced the number of breast cancer cells in proportion to the dose and time. The mechanisms of anticancer effects included cytotoxicity, apoptosis, cell targeting, gene expression regulation, and cell lysis. Summarily, bee venom and its components exert anticancer effects on human breast cancer cells. Depending on the mechanisms of anticancer effects, side effects are expected to be reduced by using various vehicles. Bee venom and its components have the potential to prevent and treat breast cancer in the future.
Collapse
Affiliation(s)
- Na-Yoen Kwon
- Department of Obstetrics and Gynecology, College of Korean Medicine, Ga-Chon University, Seongnam-si 13120, Korea;
| | - Soo-Hyun Sung
- Department of Policy Development, National Institute of Korean Medicine Development, Seoul 04554, Korea;
| | - Hyun-Kyung Sung
- Department of Korean Medicine Pediatrics, School of Korean Medicine, Semyung University, Jecheon 27136, Korea
- Correspondence: (H.-K.S.); (J.-K.P.); Tel.: +82-43-841-1739 (H.-K.S.); +82-55-360-5978 (J.-K.P.)
| | - Jang-Kyung Park
- Department of Korean Medicine Obstetrics and Gynecology, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea
- Correspondence: (H.-K.S.); (J.-K.P.); Tel.: +82-43-841-1739 (H.-K.S.); +82-55-360-5978 (J.-K.P.)
| |
Collapse
|
7
|
GPR182 limits antitumor immunity via chemokine scavenging in mouse melanoma models. Nat Commun 2022; 13:97. [PMID: 35013216 PMCID: PMC8748779 DOI: 10.1038/s41467-021-27658-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 12/03/2021] [Indexed: 01/16/2023] Open
Abstract
For many solid tumors, immune checkpoint blockade therapy has become first line treatment, yet a large proportion of patients with immunologically cold tumors do not benefit due to the paucity of tumor infiltrating lymphocytes. Here we show that the orphan G Protein-Coupled Receptor 182 (GPR182) contributes to immunotherapy resistance in cancer via scavenging chemokines that are important for lymphocyte recruitment to tumors. GPR182 is primarily upregulated in melanoma-associated lymphatic endothelial cells (LECs) during tumorigenesis, and this atypical chemokine receptor endocytoses chemokines promiscuously. In GPR182-deficient mice, T cell infiltration into transplanted melanomas increases, leading to enhanced effector T cell function and improved antitumor immunity. Ablation of GPR182 leads to increased intratumoral concentrations of multiple chemokines and thereby sensitizes poorly immunogenic tumors to immune checkpoint blockade and adoptive cellular therapies. CXCR3 blockade reverses the improved antitumor immunity and T cell infiltration characteristic of GPR182-deficient mice. Our study thus identifies GPR182 as an upstream regulator of the CXCL9/CXCL10/CXCR3 axis that limits antitumor immunity and as a potential therapeutic target in immunologically cold tumors. Immunologically cold tumours don’t respond to immune checkpoint blockade inhibition due to poor recruitment of anti-tumour T cells. Authors show here that melanoma-associated lymphatic endothelial cells express G Protein-Coupled Receptor 182 that scavenges CXCL9 and other chemokines necessary for T cell recruitment.
Collapse
|
8
|
Fleischer JR, Jodszuweit CA, Ghadimi M, De Oliveira T, Conradi LC. Vascular Heterogeneity With a Special Focus on the Hepatic Microenvironment. Front Physiol 2020; 11:591901. [PMID: 33262705 PMCID: PMC7686534 DOI: 10.3389/fphys.2020.591901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 09/28/2020] [Indexed: 12/28/2022] Open
Abstract
Utilizing single-cell sequencing, recent studies were able to analyze at a greater resolution the heterogeneity of the vasculature and its complex composition in different tissues. Differing subpopulations have been detected, distinguishable only by their transcriptome. Designed to provide further insight into the heterogeneity of the functional vascular tissue, endothelial cells have been the main target of those studies. This review aims to present a synopsis of the variability of the different vascular beds, their endothelial variety, and the supporting cells that allow the vessels to serve their various purposes. Firstly, we are going to chart vascular tissue heterogeneity on a cellular level, describing endothelial diversity as well as stromal microenvironmental variety and interaction in a physiological setting. Secondly, we will summarize the current knowledge of pathological vessel formation in the context of cancer. Conventional anti-tumor therapeutic targets as well as anti-angiogenetic therapy is frequently limited by poor response of the tumor tissue. Reasons for moderate response and resistance to treatment can be found through different drivers of angiogenesis, different mechanisms of blood supply, but also in poorly understood tissue diversity. Based on this, we are comparing how pathologies alter the normal structure of vascular tissues highlighting the involved mechanisms. Lastly, illustrating the concept above, we will focus on the hepatic microenvironment, an organ of frequent metastatic spreading (e.g., from colorectal, breast, and lung cancers). We will address how the hepatic vasculature usually develops and subsequently we will describe how common liver metastases vary in their vasculature and the way they supply themselves (e.g., angiogenesis versus vessel co-option).
Collapse
Affiliation(s)
- Johannes Robert Fleischer
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Chiara Angelina Jodszuweit
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Michael Ghadimi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago De Oliveira
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Lena-Christin Conradi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
9
|
Tumor Endothelial Cell-A Biological Tool for Translational Cancer Research. Int J Mol Sci 2020; 21:ijms21093238. [PMID: 32375250 PMCID: PMC7247330 DOI: 10.3390/ijms21093238] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/14/2022] Open
Abstract
Going from bench to bedside is a simplified description of translational research, with the ultimate goal being to improve the health status of mankind. Tumor endothelial cells (TECs) perform angiogenesis to support the growth, establishment, and dissemination of tumors to distant organs. TECs have various features that distinguish them from normal endothelial cells, which include alterations in gene expression patterns, higher angiogenic and metabolic activities, and drug resistance tendencies. The special characteristics of TECs enhance the vulnerability of tumor blood vessels toward antiangiogenic therapeutic strategies. Therefore, apart from being a viable therapeutic target, TECs would act as a better mediator between the bench (i.e., angiogenesis research) and the bedside (i.e., clinical application of drugs discovered through research). Exploitation of TEC characteristics could reveal unidentified strategies of enhancing and monitoring antiangiogenic therapy in the treatment of cancer, which are discussed in this review.
Collapse
|
10
|
Identification of Macrophage Genotype and Key Biological Pathways in Circulating Angiogenic Cell Transcriptome. Stem Cells Int 2019; 2019:9545261. [PMID: 31191690 PMCID: PMC6525806 DOI: 10.1155/2019/9545261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/14/2019] [Accepted: 02/13/2019] [Indexed: 11/17/2022] Open
Abstract
Background Circulating angiogenic cells (CAC) have been identified as important regulators of vascular biology. However, there is still considerable debate about the genotype and function of CAC. Methods and Results Data from publicly available gene expression data sets were used to analyse the transcriptome of in vitro cultured CAC (CACiv). Genes and pathways of interest were further evaluated using qPCR comparing CACiv versus CD14+ monocytic cells. The CACiv transcriptome strongly related to tissue macrophages, and more specifically to regulatory M2c macrophages. The cytokine expression profile of CACiv was predominantly immune modulatory and resembled the cytokine expression of tumor-associated macrophages (TAM). Pathway analysis revealed previously unrecognized biological processes in CACiv, such as riboflavin metabolism and liver X receptor (LXR)/retinoid X receptor (RXR) and farnesoid X receptor (FXR)/retinoid X receptor (RXR) pathways. Analysis of endothelial-specific genes did not show evidence for endothelial transdifferentiation. Conclusions CACiv are genotypically similar to regulatory M2c macrophages and lack signs of endothelial differentiation.
Collapse
|
11
|
Zmetakova I, Kalinkova L, Smolkova B, Horvathova Kajabova V, Cierna Z, Danihel L, Bohac M, Sedlackova T, Minarik G, Karaba M, Benca J, Cihova M, Buocikova V, Miklikova S, Mego M, Fridrichova I. A disintegrin and metalloprotease 23 hypermethylation predicts decreased disease-free survival in low-risk breast cancer patients. Cancer Sci 2019; 110:1695-1704. [PMID: 30815959 PMCID: PMC6500989 DOI: 10.1111/cas.13985] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/13/2019] [Accepted: 02/24/2019] [Indexed: 01/06/2023] Open
Abstract
A Disintegrin And Metalloprotease 23 (ADAM23), a member of the ADAM family, is involved in neuronal differentiation and cancer. ADAM23 is considered a possible tumor suppressor gene and is frequently downregulated in various types of malignancies. Its epigenetic silencing through promoter hypermethylation was observed in breast cancer (BC). In the present study, we evaluated the prognostic significance of ADAM23 promoter methylation for hematogenous spread and disease-free survival (DFS). Pyrosequencing was used to quantify ADAM23 methylation in tumors of 203 BC patients. Presence of circulating tumor cells (CTC) in their peripheral blood was detected by quantitative RT-PCR. Expression of epithelial (KRT19) or mesenchymal (epithelial-mesenchymal transition [EMT]-inducing transcription factors TWIST1, SNAI1, SLUG and ZEB1) mRNA transcripts was examined in CD45-depleted peripheral blood mononuclear cells. ADAM23 methylation was significantly lower in tumors of patients with the mesenchymal CTC (P = .006). It positively correlated with Ki-67 proliferation, especially in mesenchymal CTC-negative patients (P = .001). In low-risk patients, characterized by low Ki-67 and mesenchymal CTC absence, ADAM23 hypermethylation was an independent predictor of DFS (P = .006). Our results indicate that ADAM23 is likely involved in BC progression and dissemination of mesenchymal CTC. ADAM23 methylation has the potential to function as a novel prognostic marker and therapeutic target.
Collapse
Affiliation(s)
- Iveta Zmetakova
- Cancer Research InstituteBiomedical Research CenterSlovak Academy of SciencesBratislavaSlovakia
| | - Lenka Kalinkova
- Cancer Research InstituteBiomedical Research CenterSlovak Academy of SciencesBratislavaSlovakia
| | - Bozena Smolkova
- Cancer Research InstituteBiomedical Research CenterSlovak Academy of SciencesBratislavaSlovakia
| | | | - Zuzana Cierna
- Department of PathologyFaculty of MedicineComenius UniversityBratislavaSlovakia
| | - Ludovit Danihel
- Department of PathologyFaculty of MedicineComenius UniversityBratislavaSlovakia
| | - Martin Bohac
- 2nd Department of OncologyFaculty of MedicineNational Cancer InstituteComenius UniversityBratislavaSlovakia
| | - Tatiana Sedlackova
- Institute of Molecular BiomedicineFaculty of MedicineComenius UniversityBratislavaSlovakia
| | - Gabriel Minarik
- Institute of Molecular BiomedicineFaculty of MedicineComenius UniversityBratislavaSlovakia
| | - Marian Karaba
- 2nd Department of OncologyFaculty of MedicineNational Cancer InstituteComenius UniversityBratislavaSlovakia
- Department of OncosurgeryNational Cancer InstituteBratislavaSlovakia
| | - Juraj Benca
- Department of OncosurgeryNational Cancer InstituteBratislavaSlovakia
- Department of MedicineSt. Elizabeth UniversityBratislavaSlovakia
| | - Marina Cihova
- Cancer Research InstituteBiomedical Research CenterSlovak Academy of SciencesBratislavaSlovakia
| | - Verona Buocikova
- Cancer Research InstituteBiomedical Research CenterSlovak Academy of SciencesBratislavaSlovakia
| | - Svetlana Miklikova
- Cancer Research InstituteBiomedical Research CenterSlovak Academy of SciencesBratislavaSlovakia
| | - Michal Mego
- 2nd Department of OncologyFaculty of MedicineNational Cancer InstituteComenius UniversityBratislavaSlovakia
| | - Ivana Fridrichova
- Cancer Research InstituteBiomedical Research CenterSlovak Academy of SciencesBratislavaSlovakia
| |
Collapse
|
12
|
Kalinkova L, Zmetakova I, Smolkova B, Minarik G, Sedlackova T, Horvathova Kajabova V, Cierna Z, Mego M, Fridrichova I. Decreased methylation in the SNAI2 and ADAM23 genes associated with de-differentiation and haematogenous dissemination in breast cancers. BMC Cancer 2018; 18:875. [PMID: 30189837 PMCID: PMC6127923 DOI: 10.1186/s12885-018-4783-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 08/29/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND In breast cancer (BC), deregulation of DNA methylation leads to aberrant expressions and functions of key regulatory genes. In our study, we investigated the relationship between the methylation profiles of genes associated with cancer invasivity and clinico-pathological parameters. In detail, we studied differences in the methylation levels between BC patients with haematogenous and lymphogenous cancer dissemination. METHODS We analysed samples of primary tumours (PTs), lymph node metastases (LNMs) and peripheral blood cells (PBCs) from 59 patients with sporadic disseminated BC. Evaluation of the DNA methylation levels of six genes related to invasivity, ADAM23, uPA, CXCL12, TWIST1, SNAI1 and SNAI2, was performed by pyrosequencing. RESULTS Among the cancer-specific methylated genes, we found lower methylation levels of the SNAI2 gene in histologic grade 3 tumours (OR = 0.61; 95% CI, 0.39-0.97; P = 0.038) than in fully or moderately differentiated cancers. We also evaluated the methylation profiles in patients with different cancer cell dissemination statuses (positivity for circulating tumour cells (CTCs) and/or LNMs). We detected the significant association between reduced DNA methylation of ADAM23 in PTs and presence of CTCs in the peripheral blood of patients (OR = 0.45; 95% CI, 0.23-0.90; P = 0.023). CONCLUSION The relationships between the decreased methylation levels of the SNAI2 and ADAM23 genes and cancer de-differentiation and haematogenous dissemination, respectively, indicate novel functions of those genes in the invasive processes. After experimental validation of the association between the lower values of SNAI2 and ADAM23 methylation and clinical features of aggressive BCs, these methylation profiles could improve the management of metastatic disease.
Collapse
Affiliation(s)
- Lenka Kalinkova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, v.v.i., Dubravska cesta 9, 845 05, Bratislava, Slovak Republic
| | - Iveta Zmetakova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, v.v.i., Dubravska cesta 9, 845 05, Bratislava, Slovak Republic
| | - Bozena Smolkova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, v.v.i., Dubravska cesta 9, 845 05, Bratislava, Slovak Republic
| | - Gabriel Minarik
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08, Bratislava, Slovak Republic
| | - Tatiana Sedlackova
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08, Bratislava, Slovak Republic
| | - Viera Horvathova Kajabova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, v.v.i., Dubravska cesta 9, 845 05, Bratislava, Slovak Republic
| | - Zuzana Cierna
- Institute of Pathological Anatomy, Faculty of Medicine, Comenius University, University Hospital, Sasinkova 4, 811 08, Bratislava, Slovak Republic
| | - Michal Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, National Cancer Institute, Klenova 1, 83310, Bratislava, Slovak Republic
| | - Ivana Fridrichova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, v.v.i., Dubravska cesta 9, 845 05, Bratislava, Slovak Republic.
| |
Collapse
|
13
|
Abstract
Tumor endothelial cells (TEC) play an indispensible role in tumor growth and metastasis although much of the detailed mechanism still remains elusive. In this study we characterized and compared the global gene expression profiles of TECs and control ECs isolated from human breast cancerous tissues and reduction mammoplasty tissues respectively by single cell RNA sequencing (scRNA-seq). Based on the qualified scRNA-seq libraries that we made, we found that 1302 genes were differentially expressed between these two EC phenotypes. Both principal component analysis (PCA) and heat map-based hierarchical clustering separated the cancerous versus control ECs as two distinctive clusters, and MetaCore disease biomarker analysis indicated that these differentially expressed genes are highly correlated with breast neoplasm diseases. Gene Set Enrichment Analysis software (GSEA) enriched these genes to extracellular matrix (ECM) signal pathways and highlighted 127 ECM-associated genes. External validation verified some of these ECM-associated genes are not only generally overexpressed in various cancer tissues but also specifically overexpressed in colorectal cancer ECs and lymphoma ECs. In conclusion, our data demonstrated that ECM-associated genes play pivotal roles in breast cancer EC biology and some of them could serve as potential TEC biomarkers for various cancers.
Collapse
|
14
|
Wang G, Wu M, Maloneyhuss MA, Wojcik J, Durham AC, Mason NJ, Roth DB. Actionable mutations in canine hemangiosarcoma. PLoS One 2017; 12:e0188667. [PMID: 29190660 PMCID: PMC5708669 DOI: 10.1371/journal.pone.0188667] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/10/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Angiosarcomas (AS) are rare in humans, but they are a deadly subtype of soft tissue sarcoma. Discovery sequencing in AS, especially the visceral form, is hampered by the rarity of cases. Most diagnostic material exists as archival formalin fixed, paraffin embedded tissue which serves as a poor source of high quality DNA for genome-wide sequencing. We approached this problem through comparative genomics. We hypothesized that exome sequencing a histologically similar tumor, hemangiosarcoma (HSA), that occurs in approximately 50,000 dogs per year, may lead to the identification of potential oncogenic drivers and druggable targets that could also occur in angiosarcoma. METHODS Splenic hemangiosarcomas are common in dogs, which allowed us to collect a cohort of archived matched tumor and normal tissue samples suitable for whole exome sequencing. Mapping of the reads to the latest canine reference genome (Canfam3) demonstrated that >99% of the targeted exomal regions were covered, with >80% at 20X coverage and >90% at 10X coverage. RESULTS AND CONCLUSIONS Sequence analysis of 20 samples identified somatic mutations in PIK3CA, TP53, PTEN, and PLCG1, all of which correspond to well-known tumor drivers in human cancer, in more than half of the cases. In one case, we identified a mutation in PLCG1 identical to a mutation observed previously in this gene in human visceral AS. Activating PIK3CA mutations present novel therapeutic targets, and clinical trials of targeted inhibitors are underway in human cancers. Our results lay a foundation for similar clinical trials in canine HSA, enabling a precision medicine approach to this disease.
Collapse
Affiliation(s)
- Guannan Wang
- Department of Pathology and Laboratory Medicine, Raymond and Ruth Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Ming Wu
- Illumina, San Diego, CA, United States of America
| | - Martha A. Maloneyhuss
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - John Wojcik
- Department of Pathology and Laboratory Medicine, Raymond and Ruth Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Amy C. Durham
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Nicola J. Mason
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - David B. Roth
- Department of Pathology and Laboratory Medicine, Raymond and Ruth Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| |
Collapse
|
15
|
Bani M, Decio A, Giavazzi R, Ghilardi C. Contribution of tumor endothelial cells to drug resistance: anti-angiogenic tyrosine kinase inhibitors act as p-glycoprotein antagonists. Angiogenesis 2017; 20:233-241. [DOI: 10.1007/s10456-017-9549-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 03/13/2017] [Indexed: 12/12/2022]
|
16
|
Fabian KPL, Chi-Sabins N, Taylor JL, Fecek R, Weinstein A, Storkus WJ. Therapeutic efficacy of combined vaccination against tumor pericyte-associated antigens DLK1 and DLK2 in mice. Oncoimmunology 2017; 6:e1290035. [PMID: 28405524 DOI: 10.1080/2162402x.2017.1290035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/25/2017] [Accepted: 01/26/2017] [Indexed: 12/28/2022] Open
Abstract
When compared with vascular cells in normal tissues, pericytes and vascular endothelial cells (VEC) in tumor blood vessels exhibit altered morphology and epigenetic programming that leads to the expression of unique antigens that allow for differential recognition by CD8+ T cells. We have previously shown that the Notch antagonist delta-like homolog 1 (DLK1) is a tumor pericyte-associated antigen expressed in setting of melanoma and a range of carcinomas. In this report, we show that therapeutic vaccination against DLK1 in murine models results in slowed tumor growth, but also to the compensatory expression of the DLK1 homolog, DLK2, by tumor-associated pericytes. Vaccines targeting both DLK1 and DLK2 resulted in superior antitumor benefits in association with improved activation and recruitment of antigen-specific Type 1 CD8+ T cells, reduced presence of myeloid-derived suppressive cells, T regulatory cell and tumor vascular normalization. The antitumor efficacy of vaccines coordinately targeting DLK1 and DLK2 was further improved by inclusion of PD-L1 blockade, thus defining a combination immunotherapy theoretically suitable for the treatment of a broad range of solid (vascularized) cancers.
Collapse
Affiliation(s)
- Kellsye Paula L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Nina Chi-Sabins
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Jennifer L Taylor
- Department of Dermatology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Ronald Fecek
- Department of Dermatology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Aliyah Weinstein
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; The University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
17
|
Pleshkan VV, Alekseenko IV, Tyulkina DV, Kyzmich AI, Zinovyeva MV, Sverdlov ED. Fibroblast activation protein (FAP) as a possible target of an antitumor strategy. MOLECULAR GENETICS MICROBIOLOGY AND VIROLOGY 2017. [DOI: 10.3103/s0891416816030083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
18
|
Fabian KL, Storkus WJ. Immunotherapeutic Targeting of Tumor-Associated Blood Vessels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:191-211. [PMID: 29275473 DOI: 10.1007/978-3-319-67577-0_13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pathological angiogenesis occurs during tumor progression and leads in the formation of an abnormal vasculature in the tumor microenvironment (TME). The tumor vasculature is disorganized, tortuous and leaky, resulting in high interstitial pressure and hypoxia in the TME, all of which are events that support tumor growth and survival. Given the sustaining role of the tumor vasculature, it has become an increasingly attractive target for the development of anti-cancer therapies. Antibodies, tyrosine kinase inhibitors and cancer vaccines that target pro-angiogenic factors, angiogenesis-associated receptors or tumor blood vessel-associated antigens continue to be developed and tested for therapeutic efficacy. Preferred anti-angiogenic protocols include those that "normalize" the tumor-associated vasculature which reduce hypoxia and improve tumor blood perfusion, resulting in tumor cell apoptosis, decreased immunosuppression, and enhanced effector immune cell infiltration/tumoricidal action within the TME.
Collapse
Affiliation(s)
- Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Dermatology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
19
|
Ghilardi C, Silini A, Figini S, Anastasia A, Lupi M, Fruscio R, Giavazzi R, Bani MR. Trypsinogen 4 boosts tumor endothelial cells migration through proteolysis of tissue factor pathway inhibitor-2. Oncotarget 2016; 6:28389-400. [PMID: 26318044 PMCID: PMC4695067 DOI: 10.18632/oncotarget.4949] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 07/02/2015] [Indexed: 11/25/2022] Open
Abstract
Proteases contribute to cancer in many ways, including tumor vascularization and metastasis, and their pharmacological inhibition is a potential anticancer strategy. We report that human endothelial cells (EC) express the trypsinogen 4 isoform of the serine protease 3 (PRSS3), and lack both PRSS2 and PRSS1. Trypsinogen 4 expression was upregulated by the combined action of VEGF-A, FGF-2 and EGF, angiogenic factors representative of the tumor microenvironment. Suppression of trypsinogen 4 expression by siRNA inhibited the angiogenic milieu-induced migration of EC from cancer specimens (tumor-EC), but did not affect EC from normal tissues. We identified tissue factor pathway inhibitor-2 (TFPI-2), a matrix associated inhibitor of cell motility, as the functional target of trypsinogen 4, which cleaved TFPI-2 and removed it from the matrix put down by tumor-EC. Silencing tumor-EC for trypsinogen 4 accumulated TFPI2 in the matrix. Showing that angiogenic factors stimulate trypsinogen 4 expression, which hydrolyses TFPI-2 favoring a pro-migratory situation, our study suggests a new pathway linking tumor microenvironment signals to endothelial cell migration, which is essential for angiogenesis and blood vessel remodeling. Abolishing trypsinogen 4 functions might be an exploitable strategy as anticancer, particularly anti-vascular, therapy.
Collapse
Affiliation(s)
- Carmen Ghilardi
- Laboratory of Biology and Treatment of Metastases, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Antonietta Silini
- Laboratory of Biology and Treatment of Metastases, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Sara Figini
- Laboratory of Biology and Treatment of Metastases, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Alessia Anastasia
- Laboratory of Biology and Treatment of Metastases, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Monica Lupi
- Laboratory of Cancer Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Robert Fruscio
- Clinic of Obstetrics and Gynecology, University of Milan-Bicocca, San Gerardo Hospital, Monza, Italy
| | - Raffaella Giavazzi
- Laboratory of Biology and Treatment of Metastases, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Maria Rosa Bani
- Laboratory of Biology and Treatment of Metastases, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
20
|
Xiong R, Sun J, Liu K, Xu Y, He S. Effect of CPU-XT-008, a combretastatin A-4 analogue, on the proliferation, apoptosis and expression of vascular endothelial growth factor and basic fibroblast growth factor in human umbilical vein endothelial cells. Oncol Lett 2016; 11:491-499. [PMID: 26870239 DOI: 10.3892/ol.2015.3867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 09/22/2015] [Indexed: 11/05/2022] Open
Abstract
The present study investigated the effect of the combretastatin A-4 analogue CPU-XT-008 on the proliferation, apoptosis and expression of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (FGF-2) in human umbilical vein endothelial cells (HUVECs). The proliferation capacity of HUVECs was analyzed with a cell viability assay, while their apoptosis and migration abilities were evaluated via flow cytometry and monolayer denudation assay, respectively. The mRNA and protein expression levels of VEGF and FGF-2 in these cells were determined by reverse transcription-polymerase chain reaction, and cell-based ELISA, western blotting and immunocytochemistry, respectively. The results demonstrated that CPU-XT-008 inhibited proliferation and migration, and induced apoptosis in HUVECs in a dose-dependent manner. In addition, CPU-XT-008 downregulated the mRNA and protein expression levels of VEGF and FGF-2 in these cells. These findings suggest that CPU-XT-008 exerts anti-angiogenic effects in HUVECs, which may explain the inhibition of cell proliferation and migration, induction of apoptosis, and reduction in the mRNA and protein expression levels of VEGF and FGF-2 observed in the present study.
Collapse
Affiliation(s)
- Rui Xiong
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, P.R. China
| | - Jing Sun
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, P.R. China
| | - Kun Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Yungen Xu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Shuying He
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, P.R. China
| |
Collapse
|
21
|
Pleshkan VV, Alekseenko IV, Tyulkina DV, Kyzmich AI, Zinovyeva MV, Sverdlov ED. Fibroblast activation protein (FAP) as a possible target of the antitumor strategy. ACTA ACUST UNITED AC 2016. [DOI: 10.18821/0208-0613-2016-34-3-90-97] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
22
|
Thoppil RJ, Adapala RK, Cappelli HC, Kondeti V, Dudley AC, Gary Meszaros J, Paruchuri S, Thodeti CK. TRPV4 channel activation selectively inhibits tumor endothelial cell proliferation. Sci Rep 2015; 5:14257. [PMID: 26388427 PMCID: PMC4585691 DOI: 10.1038/srep14257] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 08/21/2015] [Indexed: 12/21/2022] Open
Abstract
Endothelial cell proliferation is a critical event during angiogenesis, regulated by both soluble factors and mechanical forces. Although the proliferation of tumor cells is studied extensively, little is known about the proliferation of tumor endothelial cells (TEC) and its contribution to tumor angiogenesis. We have recently shown that reduced expression of the mechanosensitive ion channel TRPV4 in TEC causes aberrant mechanosensitivity that result in abnormal angiogenesis. Here, we show that TEC display increased proliferation compared to normal endothelial cells (NEC). Further, we found that TEC exhibit high basal ERK1/2 phosphorylation and increased expression of proliferative genes important in the G1/S phase of the cell cycle. Importantly, pharmacological activation of TRPV4, with a small molecular activator GSK1016790A (GSK), significantly inhibited TEC proliferation, but had no effect on the proliferation of NEC or the tumor cells (epithelial) themselves. This reduction in TEC proliferation by TRPV4 activation was correlated with a decrease in high basal ERK1/2 phosphorylation. Finally, using a syngeneic tumor model revealed that TRPV4 activation, with GSK, significantly reduced endothelial cell proliferation in vivo. Our findings suggest that TRPV4 channels regulate tumor angiogenesis by selectively inhibiting tumor endothelial cell proliferation.
Collapse
Affiliation(s)
- Roslin J Thoppil
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272.,School of Biomedical Sciences, Kent State University, Kent, OH 44240
| | - Ravi K Adapala
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272.,School of Biomedical Sciences, Kent State University, Kent, OH 44240
| | - Holly C Cappelli
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272.,School of Biomedical Sciences, Kent State University, Kent, OH 44240
| | - Vinay Kondeti
- Department of Chemistry, University of Akron, Akron, OH 44325
| | - Andrew C Dudley
- Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, NC 27599
| | - J Gary Meszaros
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272.,School of Biomedical Sciences, Kent State University, Kent, OH 44240
| | | | - Charles K Thodeti
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272.,School of Biomedical Sciences, Kent State University, Kent, OH 44240
| |
Collapse
|
23
|
Borsotti P, Ghilardi C, Ostano P, Silini A, Dossi R, Pinessi D, Foglieni C, Scatolini M, Lacal PM, Ferrari R, Moscatelli D, Sangalli F, D'Atri S, Giavazzi R, Bani MR, Chiorino G, Taraboletti G. Thrombospondin-1 is part of a Slug-independent motility and metastatic program in cutaneous melanoma, in association with VEGFR-1 and FGF-2. Pigment Cell Melanoma Res 2014; 28:73-81. [PMID: 25256553 DOI: 10.1111/pcmr.12319] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/18/2014] [Indexed: 11/28/2022]
Abstract
Differently from most transformed cells, cutaneous melanoma expresses the pleiotropic factor thrombospondin-1 (TSP-1). Herein, we show that TSP-1 (RNA and protein), undetectable in four cultures of melanocytes and a RGP melanoma, was variously present in 13 cell lines from advanced melanomas or metastases. Moreover, microarray analysis of 55 human lesions showed higher TSP-1 expression in primary melanomas and metastases than in common and dysplastic nevi. In a functional enrichment analysis, the expression of TSP-1 correlated with motility-related genes. Accordingly, TSP-1 production was associated with melanoma cell motility in vitro and lung colonization potential in vivo. VEGF/VEGFR-1 and FGF-2, involved in melanoma progression, regulated TSP-1 production. These factors were coexpressed with TSP-1 and correlated negatively with Slug (SNAI2), a cell migration master gene implicated in melanoma metastasis. We conclude that TSP-1 cooperates with FGF-2 and VEGF/VEGFR-1 in determining melanoma invasion and metastasis, as part of a Slug-independent motility program.
Collapse
Affiliation(s)
- Patrizia Borsotti
- Tumor Angiogenesis Unit, Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Haerteis S, Krappitz A, Krappitz M, Murphy JE, Bertog M, Krueger B, Nacken R, Chung H, Hollenberg MD, Knecht W, Bunnett NW, Korbmacher C. Proteolytic activation of the human epithelial sodium channel by trypsin IV and trypsin I involves distinct cleavage sites. J Biol Chem 2014; 289:19067-78. [PMID: 24841206 DOI: 10.1074/jbc.m113.538470] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Proteolytic activation is a unique feature of the epithelial sodium channel (ENaC). However, the underlying molecular mechanisms and the physiologically relevant proteases remain to be identified. The serine protease trypsin I can activate ENaC in vitro but is unlikely to be the physiologically relevant activating protease in ENaC-expressing tissues in vivo. Herein, we investigated whether human trypsin IV, a form of trypsin that is co-expressed in several extrapancreatic epithelial cells with ENaC, can activate human ENaC. In Xenopus laevis oocytes, we monitored proteolytic activation of ENaC currents and the appearance of γENaC cleavage products at the cell surface. We demonstrated that trypsin IV and trypsin I can stimulate ENaC heterologously expressed in oocytes. ENaC cleavage and activation by trypsin IV but not by trypsin I required a critical cleavage site (Lys-189) in the extracellular domain of the γ-subunit. In contrast, channel activation by trypsin I was prevented by mutating three putative cleavage sites (Lys-168, Lys-170, and Arg-172) in addition to mutating previously described prostasin (RKRK(178)), plasmin (Lys-189), and neutrophil elastase (Val-182 and Val-193) sites. Moreover, we found that trypsin IV is expressed in human renal epithelial cells and can increase ENaC-mediated sodium transport in cultured human airway epithelial cells. Thus, trypsin IV may regulate ENaC function in epithelial tissues. Our results show, for the first time, that trypsin IV can stimulate ENaC and that trypsin IV and trypsin I activate ENaC by cleavage at distinct sites. The presence of distinct cleavage sites may be important for ENaC regulation by tissue-specific proteases.
Collapse
Affiliation(s)
- Silke Haerteis
- From the Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Waldstrasse 6, 91054 Erlangen, Germany
| | - Annabel Krappitz
- From the Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Waldstrasse 6, 91054 Erlangen, Germany
| | - Matteus Krappitz
- From the Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Waldstrasse 6, 91054 Erlangen, Germany
| | - Jane E Murphy
- the UCSF Center for the Neurobiology of Digestive Diseases, Department of Surgery, University of California, San Francisco, California
| | - Marko Bertog
- From the Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Waldstrasse 6, 91054 Erlangen, Germany
| | - Bettina Krueger
- From the Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Waldstrasse 6, 91054 Erlangen, Germany
| | - Regina Nacken
- From the Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Waldstrasse 6, 91054 Erlangen, Germany
| | - Hyunjae Chung
- the Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Morley D Hollenberg
- the Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Wolfgang Knecht
- Bioscience, CVGI iMed, AstraZeneca Research and Development, 43181 Mölndal, Sweden
| | - Nigel W Bunnett
- the Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, Victoria 3052, Australia, and the Department of Pharmacology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Christoph Korbmacher
- From the Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Waldstrasse 6, 91054 Erlangen, Germany,
| |
Collapse
|
25
|
Fiorio Pla A, Munaron L. Functional properties of ion channels and transporters in tumour vascularization. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130103. [PMID: 24493751 DOI: 10.1098/rstb.2013.0103] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Vascularization is crucial for solid tumour growth and invasion, providing metabolic support and sustaining metastatic dissemination. It is now accepted that ion channels and transporters play a significant role in driving the cancer growth at all stages. They may represent novel therapeutic, diagnostic and prognostic targets for anti-cancer therapies. On the other hand, although the expression and role of ion channels and transporters in the vascular endothelium is well recognized and subject of recent reviews, only recently has their involvement in tumour vascularization been recognized. Here, we review the current literature on ion channels and transporters directly involved in the angiogenic process. Particular interest will be focused on tumour angiogenesis in vivo as well as in the different steps that drive this process in vitro, such as endothelial cell proliferation, migration, adhesion and tubulogenesis. Moreover, we compare the 'transportome' system of tumour vascular network with the physiological one.
Collapse
Affiliation(s)
- Alessandra Fiorio Pla
- Department of Life Sciences and Systems Biology, Center for Complex Systems in Molecular Biology and Medicine (SysBioM), Nanostructured Interfaces and Surfaces Centre of Excellence (NIS), University of Torino, , Via Accademia Albertina 13, Torino 10123, Italy
| | | |
Collapse
|
26
|
Tobia C, Gariano G, De Sena G, Presta M. Zebrafish embryo as a tool to study tumor/endothelial cell cross-talk. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1371-7. [DOI: 10.1016/j.bbadis.2013.01.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/14/2013] [Accepted: 01/17/2013] [Indexed: 01/20/2023]
|
27
|
Masiero M, Simões F, Han H, Snell C, Peterkin T, Bridges E, Mangala L, Wu SY, Pradeep S, Li D, Han C, Dalton H, Lopez-Berestein G, Tuynman J, Mortensen N, Li JL, Patient R, Sood A, Banham A, Harris A, Buffa F. A core human primary tumor angiogenesis signature identifies the endothelial orphan receptor ELTD1 as a key regulator of angiogenesis. Cancer Cell 2013; 24:229-41. [PMID: 23871637 PMCID: PMC3743050 DOI: 10.1016/j.ccr.2013.06.004] [Citation(s) in RCA: 238] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 02/27/2013] [Accepted: 06/06/2013] [Indexed: 11/24/2022]
Abstract
Limited clinical benefits derived from anti-VEGF therapy have driven the identification of new targets involved in tumor angiogenesis. Here, we report an integrative meta-analysis to define the transcriptional program underlying angiogenesis in human cancer. This approach identified ELTD1, an orphan G-protein-coupled receptor whose expression is induced by VEGF/bFGF and repressed by DLL4 signaling. Extensive analysis of multiple cancer types demonstrates significant upregulation of ELTD1 in tumor-associated endothelial cells, with a higher expression correlating with favorable prognosis. Importantly, ELTD1 silencing impairs endothelial sprouting and vessel formation in vitro and in vivo, drastically reducing tumor growth and greatly improving survival. Collectively, these results provide insight into the regulation of tumor angiogenesis and highlight ELTD1 as key player in blood vessel formation.
Collapse
Affiliation(s)
- Massimo Masiero
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
- Cancer Research UK Department of Oncology, Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK
| | - Filipa Costa Simões
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Hee Dong Han
- Department of Gynecologic Oncology, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- Center for RNAi and Non-Coding RNA, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Cameron Snell
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Tessa Peterkin
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK
| | - Esther Bridges
- Cancer Research UK Department of Oncology, Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK
| | - Lingegowda S. Mangala
- Department of Gynecologic Oncology, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- Center for RNAi and Non-Coding RNA, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Sherry Yen-Yao Wu
- Department of Gynecologic Oncology, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Sunila Pradeep
- Department of Gynecologic Oncology, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Demin Li
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Cheng Han
- Cancer Research UK Department of Oncology, Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK
| | - Heather Dalton
- Department of Gynecologic Oncology, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Gabriel Lopez-Berestein
- Center for RNAi and Non-Coding RNA, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- Department of Cancer Biology, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- Department of Experimental Therapeutics, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Jurriaan B. Tuynman
- Department of Colorectal Surgery, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Neil Mortensen
- Department of Colorectal Surgery, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Ji-Liang Li
- Cancer Research UK Department of Oncology, Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK
| | - Roger Patient
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK
| | - Anil K. Sood
- Department of Gynecologic Oncology, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- Center for RNAi and Non-Coding RNA, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- Department of Cancer Biology, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Alison H. Banham
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Adrian L. Harris
- Cancer Research UK Department of Oncology, Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK
- Corresponding author
| | - Francesca M. Buffa
- Cancer Research UK Department of Oncology, Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK
- Corresponding author
| |
Collapse
|
28
|
Doig TN, Hume DA, Theocharidis T, Goodlad JR, Gregory CD, Freeman TC. Coexpression analysis of large cancer datasets provides insight into the cellular phenotypes of the tumour microenvironment. BMC Genomics 2013; 14:469. [PMID: 23845084 PMCID: PMC3721986 DOI: 10.1186/1471-2164-14-469] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 06/25/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Biopsies taken from individual tumours exhibit extensive differences in their cellular composition due to the inherent heterogeneity of cancers and vagaries of sample collection. As a result genes expressed in specific cell types, or associated with certain biological processes are detected at widely variable levels across samples in transcriptomic analyses. This heterogeneity also means that the level of expression of genes expressed specifically in a given cell type or process, will vary in line with the number of those cells within samples or activity of the pathway, and will therefore be correlated in their expression. RESULTS Using a novel 3D network-based approach we have analysed six large human cancer microarray datasets derived from more than 1,000 individuals. Based upon this analysis, and without needing to isolate the individual cells, we have defined a broad spectrum of cell-type and pathway-specific gene signatures present in cancer expression data which were also found to be largely conserved in a number of independent datasets. CONCLUSIONS The conserved signature of the tumour-associated macrophage is shown to be largely-independent of tumour cell type. All stromal cell signatures have some degree of correlation with each other, since they must all be inversely correlated with the tumour component. However, viewed in the context of established tumours, the interactions between stromal components appear to be multifactorial given the level of one component e.g. vasculature, does not correlate tightly with another, such as the macrophage.
Collapse
Affiliation(s)
- Tamasin N Doig
- Centre for Inflammation Research, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | | | | | | | | | | |
Collapse
|
29
|
Maric G, Rose AA, Annis MG, Siegel PM. Glycoprotein non-metastatic b (GPNMB): A metastatic mediator and emerging therapeutic target in cancer. Onco Targets Ther 2013; 6:839-52. [PMID: 23874106 PMCID: PMC3711880 DOI: 10.2147/ott.s44906] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Molecularly targeted therapies are rapidly growing with respect to their clinical development and impact on cancer treatment due to their highly selective anti-tumor action. However, many aggressive cancers such as triple-negative breast cancer (TNBC) currently lack well-defined therapeutic targets against which such agents can be developed. The identification of tumor-associated antigens and the generation of antibody drug-conjugates represent an emerging area of intense interest and growth in the field of cancer therapeutics. Glycoprotein non-metastatic b (GPNMB) has recently been identified as a gene that is over-expressed in numerous cancers, including TNBC, and often correlates with the metastatic phenotype. In breast cancer, GPNMB expression in the tumor epithelium is associated with a reduction in disease-free and overall survival. Based on these findings, glembatumumab vedotin (CDX-011), an antibody-drug conjugate that selectively targets GPNMB, is currently being investigated in clinical trials for patients with metastatic breast cancer and unresectable melanoma. This review discusses the physiological and potential pathological roles of GPNMB in normal and cancer tissues, respectively, and details the clinical advances and challenges in targeting GPNMB-expressing malignancies.
Collapse
Affiliation(s)
- Gordana Maric
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada ; Department of Medicine, McGill University, Montréal, Québec, Canada
| | | | | | | |
Collapse
|
30
|
Tsui KH, Chang YL, Feng TH, Chang PL, Juang HH. Glycoprotein transmembrane nmb: an androgen-downregulated gene attenuates cell invasion and tumorigenesis in prostate carcinoma cells. Prostate 2012; 72:1431-42. [PMID: 22290289 DOI: 10.1002/pros.22494] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 01/02/2012] [Indexed: 12/22/2022]
Abstract
BACKGROUND Glycoprotein transmembrane nmb (GPNMB) gene was originally identified in osteoblasts and belongs to the pmel-17/nmb family. The function or regulation of GPNMB in the human prostate remains unknown. METHODS The expression of GPNMB in prostate carcinoma cells were determined by real-time reverse transcription-polymerase chain reaction (RT-qPCR) and immunoblot assays. Effects of ectopic GPNMB overexpression on cell proliferation, invasion, and tumorigenesis were determined by (3) H-thymidine incorporation, matrigel invasion, soft agar cloning assays, and murine xenograft study. Effects of GPNMB, p53, and androgen on target gene were assessed using RT-PCR, immunoblotting, and transient gene expression assays. RESULTS In vitro analysis using several prostate cell lines suggested that expression of GPNMB may be relevant to the extent of neoplasia. Ectopic overexpression of GPNMB significantly attenuated cell proliferation and invasion and exerted antitumorigenic activity on PC-3 cells in vitro and in vivo. GPNMB overexpression induced the gene expressions of N-myc downstream regulated gene 1 (Ndrg1) and maspin in PC-3 cells. Doxorubicin treatment or transient overexpression of p53 increased GPNMB expression. Androgen (R1881) treatment has a divergent effect on gene expression of prostate-specific antigen (PSA) and GPNMB in LNCaP cells. Androgen treatment enhanced cell proliferation but downregulated GPNMB protein expression in stably overexpressed androgen receptor (AR) CA-HPV-10 cells. CONCLUSIONS Together these results suggest that GPNMB gene is a p53- and androgen-dysregulated gene and should be regarded as an anti-tumor gene for prostate cancer. The enhancement of Ndrg1 and maspin gene expressions may account for the anti-proliferative and anti-invasive function of GPNMB in PC-3 cells.
Collapse
Affiliation(s)
- Ke-Hung Tsui
- Department of Urology, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan, Taiwan, ROC
| | | | | | | | | |
Collapse
|
31
|
Neagu M, Albulescu R, Tanase C. Signaling molecule c-Met is a biomarker for favorable prognostic in Hodgkin lymphoma. Biomark Med 2012; 6:199-200. [PMID: 22448794 DOI: 10.2217/bmm.12.5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Monica Neagu
- "Victor Babes" National Institute of Pathology, Bucharest, Romania.
| | | | | |
Collapse
|
32
|
Zhao X, Bose A, Komita H, Taylor JL, Chi N, Lowe DB, Okada H, Cao Y, Mukhopadhyay D, Cohen PA, Storkus WJ. Vaccines targeting tumor blood vessel antigens promote CD8(+) T cell-dependent tumor eradication or dormancy in HLA-A2 transgenic mice. THE JOURNAL OF IMMUNOLOGY 2012; 188:1782-8. [PMID: 22246626 DOI: 10.4049/jimmunol.1101644] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have recently shown that effective cytokine gene therapy of solid tumors in HLA-A2 transgenic (HHD) mice lacking murine MHC class I molecule expression results in the generation of HLA-A2-restricted CD8(+) T effector cells selectively recognizing tumor blood vessel-associated pericytes and/or vascular endothelial cells. Using an HHD model in which HLA-A2(neg) tumor (MC38 colon carcinoma or B16 melanoma) cells are not recognized by the CD8(+) T cell repertoire, we now show that vaccines on the basis of tumor-associated blood vessel Ags (TBVA) elicit protective Tc1-dependent immunity capable of mediating tumor regression or extending overall survival. Vaccine efficacy was not observed if (HLA-A2(neg)) wild-type C57BL/6 mice were instead used as recipient animals. In the HHD model, effective vaccination resulted in profound infiltration of tumor lesions by CD8(+) (but not CD4(+)) T cells, in a coordinate reduction of CD31(+) blood vessels in the tumor microenvironment, and in the "spreading" of CD8(+) T cell responses to alternate TBVA that were not intrinsic to the vaccine. Protective Tc1-mediated immunity was durable and directly recognized pericytes and/or vascular endothelial cells flow-sorted from tumor tissue but not from tumor-uninvolved normal kidneys harvested from these same animals. Strikingly, the depletion of CD8(+), but not CD4(+), T cells at late time points after effective therapy frequently resulted in the recurrence of disease at the site of the regressed primary lesion. This suggests that the vaccine-induced anti-TBVA T cell repertoire can mediate the clinically preferred outcomes of either effectively eradicating tumors or policing a state of (occult) tumor dormancy.
Collapse
Affiliation(s)
- Xi Zhao
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
|
34
|
Regulator of G-protein signaling 5 (RGS5) protein: a novel marker of cancer vasculature elicited and sustained by the tumor's proangiogenic microenvironment. Cell Mol Life Sci 2011; 69:1167-78. [PMID: 22130514 PMCID: PMC3299962 DOI: 10.1007/s00018-011-0862-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 10/10/2011] [Accepted: 10/11/2011] [Indexed: 01/26/2023]
Abstract
We previously identified regulator of G-protein signaling 5 (RGS5) among several genes expressed by tumor-derived endothelial cells (EC). In this study, we provide the first in vivo/ex vivo evidence of RGS5 protein in the vasculature of ovarian carcinoma clinical specimens and its absence in human ovaries. Consistent with this, we show higher amounts of Rgs5 transcript in EC isolated from human cancers (as opposed to normal tissues) and demonstrate that expression is sustained by a milieu of factors typical of the proangiogenic tumor environment, including vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (FGF-2). Supporting these findings, we show elevated levels of Rgs5 mRNA in the stroma from strongly (as opposed to weakly) angiogenic ovarian carcinoma xenografts and accordingly, we also show more of the protein associated to the abnormal vasculature. RGS5 protein predominantly colocalizes with the endothelium expressing platelet/endothelial cell adhesion molecule-1 (PECAM-1/CD31) and to a much lesser extent with perivascular/mural cells expressing platelet-derived growth factor receptor-beta (PDGFR-β) or alpha smooth muscle actin (αSMA). To toughen the relevance of the findings, we demonstrate RGS5 in the blood vessels of other cancer models endowed with a proangiogenic environment, such as human melanoma and renal carcinoma xenografts; to the contrary, it was undetectable in the vasculature of normal mouse tissues. RGS5 expression by the cancer vasculature triggered and retained by the proangiogenic microenvironment supports its exploitation as a novel biomarker and opens the path to explore new possibilities of therapeutic intervention aimed at targeting tumor blood vessels.
Collapse
|
35
|
Sefcik LS, Wilson JL, Papin JA, Botchwey EA. Harnessing systems biology approaches to engineer functional microvascular networks. TISSUE ENGINEERING PART B-REVIEWS 2010; 16:361-70. [PMID: 20121415 DOI: 10.1089/ten.teb.2009.0611] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Microvascular remodeling is a complex process that includes many cell types and molecular signals. Despite a continued growth in the understanding of signaling pathways involved in the formation and maturation of new blood vessels, approximately half of all compounds entering clinical trials will fail, resulting in the loss of much time, money, and resources. Most pro-angiogenic clinical trials to date have focused on increasing neovascularization via the delivery of a single growth factor or gene. Alternatively, a focus on the concerted regulation of whole networks of genes may lead to greater insight into the underlying physiology since the coordinated response is greater than the sum of its parts. Systems biology offers a comprehensive network view of the processes of angiogenesis and arteriogenesis that might enable the prediction of drug targets and whether or not activation of the targets elicits the desired outcome. Systems biology integrates complex biological data from a variety of experimental sources (-omics) and analyzes how the interactions of the system components can give rise to the function and behavior of that system. This review focuses on how systems biology approaches have been applied to microvascular growth and remodeling, and how network analysis tools can be utilized to aid novel pro-angiogenic drug discovery.
Collapse
Affiliation(s)
- Lauren S Sefcik
- Department of Chemical and Biomolecular Engineering, Lafayette College, Easton, Pennsylvania, USA
| | | | | | | |
Collapse
|
36
|
Rose AAN, Annis MG, Dong Z, Pepin F, Hallett M, Park M, Siegel PM. ADAM10 releases a soluble form of the GPNMB/Osteoactivin extracellular domain with angiogenic properties. PLoS One 2010; 5:e12093. [PMID: 20711474 PMCID: PMC2919417 DOI: 10.1371/journal.pone.0012093] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 07/18/2010] [Indexed: 01/10/2023] Open
Abstract
Background Glycoprotein non-metastatic melanoma protein B (GPNMB)/Osteoactivin (OA) is a transmembrane protein expressed in approximately 40–75% of breast cancers. GPNMB/OA promotes the migration, invasion and metastasis of breast cancer cells; it is commonly expressed in basal/triple-negative breast tumors and is associated with shorter recurrence-free and overall survival times in patients with breast cancer. Thus, GPNMB/OA represents an attractive target for therapeutic intervention in breast cancer; however, little is known about the functions of GPNMB/OA within the primary tumor microenvironment. Methodology/Principal Findings We have employed mouse and human breast cancer cells to investigate the effects of GPNMB/OA on tumor growth and angiogenesis. GPNMB/OA-expressing tumors display elevated endothelial recruitment and reduced apoptosis when compared to vector control-derived tumors. Primary human breast cancers characterized by high vascular density also display elevated levels of GPNMB/OA when compared to those with low vascular density. Using immunoblot and ELISA assays, we demonstrate the GPNMB/OA ectodomain is shed from the surface of breast cancer cells. Transient siRNA-mediated knockdown studies of known sheddases identified ADAM10 as the protease responsible for GPNMB/OA processing. Finally, we demonstrate that the shed extracellular domain (ECD) of GPNMB/OA can promote endothelial migration in vitro. Conclusions/Significance GPNMB/OA expression promotes tumor growth, which is associated with enhanced endothelial recruitment. We identify ADAM10 as a sheddase capable of releasing the GPNMB/OA ectodomain from the surface of breast cancer cells, which induces endothelial cell migration. Thus, ectodomain shedding may serve as a novel mechanism by which GPNMB/OA promotes angiogenesis in breast cancer.
Collapse
Affiliation(s)
- April A. N. Rose
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Matthew G. Annis
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Zhifeng Dong
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Francois Pepin
- Life Science Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Michael Hallett
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- McGill Centre for Bioinformatics, McGill University, Montreal, Quebec, Canada
| | - Morag Park
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Peter M. Siegel
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
37
|
|
38
|
Rose AAN, Grosset AA, Dong Z, Russo C, Macdonald PA, Bertos NR, St-Pierre Y, Simantov R, Hallett M, Park M, Gaboury L, Siegel PM. Glycoprotein nonmetastatic B is an independent prognostic indicator of recurrence and a novel therapeutic target in breast cancer. Clin Cancer Res 2010; 16:2147-56. [PMID: 20215530 DOI: 10.1158/1078-0432.ccr-09-1611] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE Although the murine orthologue of glycoprotein nonmetastatic B (GPNMB), Osteoactivin, promotes breast cancer metastasis in an in vivo mouse model, its importance in human breast cancer is unknown. We have examined the significance of GPNMB expression as a prognostic indicator of recurrence and assessed its potential as a novel therapeutic target in breast cancer. EXPERIMENTAL DESIGN The clinical significance of GPNMB expression in breast cancer was addressed by analyzing GPNMB levels in several published gene expression data sets and two independent tissue microarrays derived from human breast tumors. GPNMB-expressing human breast cancer cell lines were further used to validate a toxin-conjugated anti-GPNMB antibody as a novel therapeutic agent. RESULTS GPNMB expression correlates with shorter recurrence times and reduced overall survival of breast cancer patients. Epithelial-specific GPNMB staining is an independent prognostic indicator for breast cancer recurrence. GPNMB is highly expressed in basal and triple-negative breast cancers and is associated with increased risk of recurrence within this subtype. GPNMB expression confers a more migratory and invasive phenotype on breast cancer cells and sensitizes them to killing by CDX-011 (glembatumumab vedotin), a GPNMB-targeted antibody-drug conjugate. CONCLUSIONS GPNMB expression is associated with the basal/triple-negative subtype and is a prognostic marker of poor outcome in patients with breast cancer. CDX-011 (glembatumumab vedotin) is a promising new targeted therapy for patients with metastatic triple-negative breast cancers, a patient population that currently lacks targeted-therapy options.
Collapse
Affiliation(s)
- April A N Rose
- Department of Medicine, Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West Montréal, Québec, Canada, H3A 1A3
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Silini A, Ghilardi C, Ardinghi C, Bernasconi S, Oliva P, Carraro F, Naldini A, Bani MR, Giavazzi R. Protease-activated receptor-1 (PAR-1) promotes the motility of human melanomas and is associated to their metastatic phenotype. Clin Exp Metastasis 2009; 27:43-53. [PMID: 20084489 DOI: 10.1007/s10585-009-9301-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Accepted: 12/03/2009] [Indexed: 10/20/2022]
Abstract
Protease-activated receptor-1 (PAR-1) is a unique G-protein-coupled receptor belonging to the protease-activated receptor family. Its activation leads to downstream signaling events that launch a variety of cellular responses related to tumor progression. PAR-1 expression has been associated to a variety of human cancers, and our previous studies reveal a high PAR-1 expression in melanoma specimens as compared to common nevi. In the present study, we investigated the contribution of PAR-1 to the malignant phenotype of human melanoma cell lines obtained from cutaneous primary lesions, capable of different metastatic behaviors in the patients from which they have been derived. We found that melanoma cells isolated from lesions giving rise to metastases in patients (WM115, WM278A, WM1361A, WM983A), had higher PAR-1 mRNA and protein expression, as compared to those obtained from lesions that did not develop metastatic disease (WM793, WM35). The cells isolated from metastatic primary lesions were able to colonize the lungs of immunodeficient SCID mice while those isolated from non-metastatic lesions were not. Additionally, cells expressing elevated PAR-1 had higher migratory and invasive abilities than those holding minimal PAR-1 expression. The migration and invasion capabilities of the melanoma cells expressing high PAR-1 were hampered by genetic and pharmacological interventions. The reduction of PAR-1 expression by siRNA and the inhibition of PAR-1 function by the SCH79797 specific antagonist significantly decreased the melanoma cell motility and invasiveness, down to an extent similar to that of the non-metastatic and low PAR-1 expressing cells. Our results provide strong evidence supporting the implication of PAR-1 in the malignant progression of human melanoma.
Collapse
Affiliation(s)
- Antonietta Silini
- Laboratory of Biology and Treatment of Metastases, Department of Oncology, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Javerzat S, Franco M, Herbert J, Platonova N, Peille AL, Pantesco V, De Vos J, Assou S, Bicknell R, Bikfalvi A, Hagedorn M. Correlating global gene regulation to angiogenesis in the developing chick extra-embryonic vascular system. PLoS One 2009; 4:e7856. [PMID: 19924294 PMCID: PMC2774277 DOI: 10.1371/journal.pone.0007856] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 10/17/2009] [Indexed: 11/18/2022] Open
Abstract
Background Formation of blood vessels requires the concerted regulation of an unknown number of genes in a spatial-, time- and dosage-dependent manner. Determining genes, which drive vascular maturation is crucial for the identification of new therapeutic targets against pathological angiogenesis. Methology/Principal Findings We accessed global gene regulation throughout maturation of the chick chorio-allantoic membrane (CAM), a highly vascularized tissue, using pan genomic microarrays. Seven percent of analyzed genes showed a significant change in expression (>2-fold, FDR<5%) with a peak occurring from E7 to E10, when key morphogenetic and angiogenic genes such as BMP4, SMO, HOXA3, EPAS1 and FGFR2 were upregulated, reflecting the state of an activated endothelium. At later stages, a general decrease in gene expression occurs, including genes encoding mitotic factors or angiogenic mediators such as CYR61, EPAS1, MDK and MYC. We identified putative human orthologs for 77% of significantly regulated genes and determined endothelial cell enrichment for 20% of the orthologs in silico. Vascular expression of several genes including ENC1, FSTL1, JAM2, LDB2, LIMS1, PARVB, PDE3A, PRCP, PTRF and ST6GAL1 was demonstrated by in situ hybridization. Up to 9% of the CAM genes were also overexpressed in human organs with related functions, such as placenta and lung or the thyroid. 21–66% of CAM genes enriched in endothelial cells were deregulated in several human cancer types (P<.0001). Interfering with PARVB (encoding parvin, beta) function profoundly changed human endothelial cell shape, motility and tubulogenesis, suggesting an important role of this gene in the angiogenic process. Conclusions/Significance Our study underlines the complexity of gene regulation in a highly vascularized organ during development. We identified a restricted number of novel genes enriched in the endothelium of different species and tissues, which may play crucial roles in normal and pathological angiogenesis.
Collapse
Affiliation(s)
- Sophie Javerzat
- INSERM U920, Laboratoire des Mécanismes Moléculaires de l'Angiogenèse, Université Bordeaux 1, Talence, France
- Université Bordeaux 1, Talence, France
| | - Mélanie Franco
- INSERM U920, Laboratoire des Mécanismes Moléculaires de l'Angiogenèse, Université Bordeaux 1, Talence, France
- Université Bordeaux 1, Talence, France
- * E-mail:
| | - John Herbert
- Molecular Angiogenesis Group, Institute of Biomedical Research, University of Birmingham, Medical School, Birmingham, United Kingdom
| | - Natalia Platonova
- INSERM U920, Laboratoire des Mécanismes Moléculaires de l'Angiogenèse, Université Bordeaux 1, Talence, France
- Université Bordeaux 1, Talence, France
| | - Anne-Lise Peille
- INSERM U920, Laboratoire des Mécanismes Moléculaires de l'Angiogenèse, Université Bordeaux 1, Talence, France
- Université Bordeaux 1, Talence, France
| | - Véronique Pantesco
- Institut de Recherche en Biothérapie, Hôpital Saint-Eloi, CHU de Montpellier, Montpellier, France
| | - John De Vos
- Institut de Recherche en Biothérapie, Hôpital Saint-Eloi, CHU de Montpellier, Montpellier, France
| | - Said Assou
- Institut de Recherche en Biothérapie, Hôpital Saint-Eloi, CHU de Montpellier, Montpellier, France
| | - Roy Bicknell
- Molecular Angiogenesis Group, Institute of Biomedical Research, University of Birmingham, Medical School, Birmingham, United Kingdom
| | - Andreas Bikfalvi
- INSERM U920, Laboratoire des Mécanismes Moléculaires de l'Angiogenèse, Université Bordeaux 1, Talence, France
- Université Bordeaux 1, Talence, France
| | - Martin Hagedorn
- INSERM U920, Laboratoire des Mécanismes Moléculaires de l'Angiogenèse, Université Bordeaux 1, Talence, France
- Université Bordeaux 1, Talence, France
- * E-mail:
| |
Collapse
|
41
|
Costa MD, Paludo KS, Klassen G, Lopes MH, Mercadante AF, Martins VR, Camargo AA, Nakao LS, Zanata SM. Characterization of a specific interaction between ADAM23 and cellular prion protein. Neurosci Lett 2009; 461:16-20. [DOI: 10.1016/j.neulet.2009.05.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Revised: 05/11/2009] [Accepted: 05/20/2009] [Indexed: 02/04/2023]
|
42
|
Dong Z, Nör JE. Transcriptional targeting of tumor endothelial cells for gene therapy. Adv Drug Deliv Rev 2009; 61:542-53. [PMID: 19393703 DOI: 10.1016/j.addr.2009.02.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Accepted: 02/05/2009] [Indexed: 12/21/2022]
Abstract
It is well known that angiogenesis plays a critical role in the pathobiology of tumors. Recent clinical trials have shown that inhibition of angiogenesis can be an effective therapeutic strategy for patients with cancer. However, one of the outstanding issues in anti-angiogenic treatment for cancer is the development of toxicities related to off-target effects of drugs. Transcriptional targeting of tumor endothelial cells involves the use of specific promoters for selective expression of therapeutic genes in the endothelial cells lining the blood vessels of tumors. Recently, several genes that are expressed specifically in tumor-associated endothelial cells have been identified and characterized. These discoveries have enhanced the prospectus of transcriptionally targeting tumor endothelial cells for cancer gene therapy. In this manuscript, we review the promoters, vectors, and therapeutic genes that have been used for transcriptional targeting of tumor endothelial cells, and discuss the prospects of such approaches for cancer gene therapy.
Collapse
Affiliation(s)
- Zhihong Dong
- Angiogenesis Research Laboratory, Department of Restorative Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
43
|
LeBeau AM, Brennen WN, Aggarwal S, Denmeade SR. Targeting the cancer stroma with a fibroblast activation protein-activated promelittin protoxin. Mol Cancer Ther 2009; 8:1378-86. [PMID: 19417147 DOI: 10.1158/1535-7163.mct-08-1170] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Fibroblast-Activation Protein-α (FAP) is a membrane-bound serine protease that is expressed on the surface of reactive stromal fibroblasts present within the majority of human epithelial tumors but is not expressed by normal tissues. FAP is a postprolyl peptidase that differs from other dipeptidyl prolyl peptidases such as diprolylpeptidase 4 in that it also has gelatinase and collagenase endopeptidase activity. Therefore, FAP represents a potential pan-tumor target whose enzymatic activity can be exploited for the intratumoral activation of prodrugs and protoxins. To evaluate FAP as a tumor-specific target, putative FAP-selective peptide protoxins were constructed through modification of the prodomain of melittin, a 26 amino acid amphipathic cytolytic peptide that is the main toxic component in the venom of the common European honeybee Apis milefera. Melittin is synthesized as promelittin, containing a 22 amino acid NH(2)-terminal prodomain rich in the amino acids proline and alanine. In this study, peptides containing truncated melittin prodomain sequences were tested on erythrocytes to determine the optimal prodomain length for inhibiting cytolytic activity. Once optimized, modified promelittin peptides were generated in which previously identified FAP substrate sequences were introduced into the prodomain. Peptide protoxins were identified that were efficiently activated by FAP and selectively toxic to FAP-expressing cell lines with an IC(50) value in the low micromolar range that is similar to melittin. Intratumoral injection of an FAP-activated protoxin produced significant lysis and growth inhibition of human breast and prostate cancer xenografts with minimal toxicity to the host animal.
Collapse
Affiliation(s)
- Aaron M LeBeau
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore Maryland 21231, USA
| | | | | | | |
Collapse
|
44
|
Munaron L, Tomatis C, Fiorio Pla A. The secret marriage between calcium and tumor angiogenesis. Technol Cancer Res Treat 2008; 7:335-9. [PMID: 18642972 DOI: 10.1177/153303460800700408] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Endothelial cell biochemistry and responsiveness to a wide variety of external stimula is regulated by intracellular calcium concentration. During the last twenty years, electrophysiology and functional imaging based on the use of fluorescent probes provided several informations about the dynamics and role of calcium at the single cell level: highly diverse extracellular agonists, such as proangiogenic growth factors and vasoactive compounds, trigger increases in intracellular calcium and specific informations are transduced for proliferation, differentiation, death, movement in physiological and pathological conditions. Obviously, the investigation at multicellular and tissutal levels is much more complex. In this review we discuss the potential specific roles of calcium signaling in tumor angiogenesis progression trying to address two key questions: (i) how can this ion play specific roles in the angiogenesis regulation; and (ii) could it be used as a target to interfere with or prevent tumor vascularization?
Collapse
Affiliation(s)
- Luca Munaron
- Department of Animal and Human Biology, University of Turin, Italy.
| | | | | |
Collapse
|