1
|
Extracellular Vesicles—A New Potential Player in the Immunology of Renal Cell Carcinoma. J Pers Med 2022; 12:jpm12050772. [PMID: 35629194 PMCID: PMC9144962 DOI: 10.3390/jpm12050772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 02/08/2023] Open
Abstract
The incidence of renal cell carcinoma (RCC) has doubled in the developed world within the last fifty years, and now it is responsible for 2–3% of diagnosed cancers. The delay in diagnosis and the not fully understood pathogenesis are the main challenges that have to be overcome. It seems that extracellular vesicles (EVs) are one of the key players in tumor development since they ensure a proper microenvironment for the tumor cells. The stimulation of angiogenesis and immunosuppression is mediated by molecules contained in EVs. It was shown that EVs derived from cancer cells can inhibit T cell proliferation, natural killer lymphocyte activation, and dendritic cell maturation by this mechanism. Moreover, EVs may be a biomarker for the response to anti-cancer treatment. In this review, we sum up the knowledge about the role of EVs in RCC pathogenesis and show their future perspectives in this field.
Collapse
|
2
|
Müller GA. Membrane insertion and intercellular transfer of glycosylphosphatidylinositol-anchored proteins: potential therapeutic applications. Arch Physiol Biochem 2020; 126:139-156. [PMID: 30445857 DOI: 10.1080/13813455.2018.1498904] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Anchorage of a subset of cell surface proteins in eukaryotic cells is mediated by a glycosylphosphatidylinositol (GPI) moiety covalently attached to the carboxy-terminus of the protein moiety. Experimental evidence for the potential of GPI-anchored proteins (GPI-AP) of being released from cells into the extracellular environment has been accumulating, which involves either the loss or retention of the GPI anchor. Release of GPI-AP from donor cells may occur spontaneously or in response to endogenous or environmental signals. The experimental evidence for direct insertion of exogenous GPI-AP equipped with the complete anchor structure into the outer plasma membrane bilayer leaflets of acceptor cells is reviewed as well as the potential underlying molecular mechanisms. Furthermore, promiscuous transfer of certain GPI-AP between plasma membranes of different cells in vivo under certain (patho)physiological conditions has been reported. Engineering of target cell surfaces using chimeric GPI-AP with complete GPI anchor may be useful for therapeutic applications.
Collapse
Affiliation(s)
- Günter A Müller
- Helmholtz Diabetes Center (HDC) at the Helmholtz Center München, Institute for Diabetes and Obesity, Oberschleissheim, Germany
- Department Biology I, Genetics, Ludwig-Maximilians-University München, Planegg-Martinsried, Germany
| |
Collapse
|
3
|
Abstract
The ability to rationally manipulate and augment the cytoplasmic membrane can be used to overcome many of the challenges faced by conventional cellular therapies and provide innovative opportunities when combined with new biotechnologies. The focus of this review is on emerging strategies used in cell functionalization, highlighting both pioneering approaches and recent developments. These will be discussed within the context of future directions in this rapidly evolving field.
Collapse
Affiliation(s)
- James Pk Armstrong
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Adam W Perriman
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK Centre for Organized Matter Chemistry and Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| |
Collapse
|
4
|
Heider S, Dangerfield JA, Metzner C. Biomedical applications of glycosylphosphatidylinositol-anchored proteins. J Lipid Res 2016; 57:1778-1788. [PMID: 27542385 PMCID: PMC5036375 DOI: 10.1194/jlr.r070201] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Indexed: 01/13/2023] Open
Abstract
Glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-APs) use a unique posttranslational modification to link proteins to lipid bilayer membranes. The anchoring structure consists of both a lipid and carbohydrate portion and is highly conserved in eukaryotic organisms regarding its basic characteristics, yet highly variable in its molecular details. The strong membrane targeting property has made the anchors an interesting tool for biotechnological modification of lipid membrane-covered entities from cells through extracellular vesicles to enveloped virus particles. In this review, we will take a closer look at the mechanisms and fields of application for GPI-APs in lipid bilayer membrane engineering and discuss their advantages and disadvantages for biomedicine.
Collapse
Affiliation(s)
- Susanne Heider
- Institute of Virology, University of Veterinary Medicine, 1210 Vienna, Austria
| | | | - Christoph Metzner
- Institute of Virology, University of Veterinary Medicine, 1210 Vienna, Austria.
| |
Collapse
|
5
|
Djafarzadeh R, Conrad C, Notohamiprodjo S, Hipp S, Niess H, Bruns CJ, Nelson PJ. Cell surface engineering using glycosylphosphatidylinositol anchored tissue inhibitor of matrix metalloproteinase-1 stimulates cutaneous wound healing. Wound Repair Regen 2014; 22:70-6. [PMID: 24393154 DOI: 10.1111/wrr.12132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 10/10/2013] [Indexed: 11/27/2022]
Abstract
The balance between matrix metalloproteinases and their endogenous tissue inhibitors (TIMPs) is an important component in effective wound healing. The biologic action of these proteins is linked in part to the stoichiometry of TIMP/matrix metalloproteinases/surface protein interactions. We recently described the effect of a glycosylphosphatidylinositol (GPI) anchored version of TIMP-1 on dermal fibroblast biology. Here, cell proliferation assays, in vitro wound healing, electrical wound, and impedance measurements were used to characterize effects of TIMP-1-GPI treatment on primary human epidermal keratinocytes. TIMP-1-GPI stimulated keratinocyte proliferation, as well as mobilization and migration. In parallel, it suppressed the migration and matrix secretion of dermal myofibroblasts, and reduced their secretion of active TGF-β1. Topical application of TIMP-1-GPI in an in vivo excisional wound model increased the rate of wound healing. The agent positively influenced different aspects of wound healing depending on the cell type studied. TIMP-1-GPI counters potential negative effects of overactive myofibroblasts and enhances the mobilization and proliferation of keratinocytes essential for effective wound healing. The application of TIMP-1-GPI represents a novel and practical clinical solution for facilitating healing of difficult wounds.
Collapse
Affiliation(s)
- Roghieh Djafarzadeh
- Medical Clinic and Outpatient Clinic IV, University of Munich, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
6
|
Gassenmaier M, Chen D, Buchner A, Henkel L, Schiemann M, Mack B, Schendel DJ, Zimmermann W, Pohla H. CXC chemokine receptor 4 is essential for maintenance of renal cell carcinoma-initiating cells and predicts metastasis. Stem Cells 2014; 31:1467-76. [PMID: 23630186 DOI: 10.1002/stem.1407] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 03/28/2013] [Indexed: 12/24/2022]
Abstract
In many solid tumors, cancer stem cells (CSC) represent a population with tumor-initiating, self-renewal, and differentiation potential, which can be identified by surface protein markers. No generally applicable markers are yet known for renal cell carcinoma (RCC). Two RCC cell lines (RCC-26, RCC-53) were found to differ widely in their capacity to form spheres in vitro and to establish tumors in mice, potentially reflecting differences in CSC content. A subpopulation expressing the CXC chemokine receptor 4 (CXCR4) was present only in the more tumorigenic cell line RCC-53. When grown as spheres, most of the RCC-53 cells were CXCR4-positive, expressed stem cell-associated transcription factor genes at elevated levels, and were more resistant toward the tyrosine kinase inhibitors sunitinib, sorafenib, and pazopanib. Sorted CXCR4-positive cells exhibited greater capacity for sphere formation and tumor growth-inducing potential in vivo than CXCR4-negative cells. Significantly, higher CXCR4 mRNA levels in primary RCC tumors from patients with localized but not disseminated disease predicted shorter survival. Downregulation of CXCR4 expression by small interfering RNA (siRNA) or pharmacological inhibition by AMD3100 compromised tumor sphere formation, viability of CXCR4-positive cells, and increased their responsiveness toward tyrosine kinase inhibitors. In conclusion, CXCR4 identifies a subpopulation of tumor-initiating cells in RCC cell lines and plays a role in their maintenance. The relative insensitivity of such cells to tyrosine kinase inhibitors might contribute to the development of therapy resistance in RCC patients. Future therapies therefore could combine blockade of the CXCR4 signaling pathway with standard therapies for more effective treatments of metastatic RCC.
Collapse
Affiliation(s)
- Maximilian Gassenmaier
- Tumor Immunology Laboratory, LIFE Center, Ludwig-Maximilians-Universität, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Khan MI, Czarnecka AM, Duchnowska R, Kukwa W, Szczylik C. Metastasis-Initiating Cells in Renal Cancer. ACTA ACUST UNITED AC 2014; 8:240-246. [PMID: 25152705 PMCID: PMC4141324 DOI: 10.2174/1574362409666140206222431] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 11/27/2013] [Accepted: 01/29/2014] [Indexed: 02/07/2023]
Abstract
Metastasis is a complex process that propagates cells from the primary or initial site of the cancer occurrence to distant parts of the body. Cancer cells break from the cancer site and circulate through the bloodstream or lymph vessels, allowing them to reach nearly all parts of the body. These circulating tumour cells (CTCs) contain specialized metastasis-initiating cells (MICs) that reside in the biological heterogeneous primary tumour. Researchers have hypothesized that metastasis of renal cell carcinoma is initiated by circulation of MICs in patients’ blood and bone marrow. Based on the cancer stem/progenitor cell concept of carcinogenesis, understanding the molecular phenotypes of metastasis-initiating cells (MICs) in renal cancer could play a vital role in developing strategies for therapeutic interventions in renal cancer. Existence of MICs among CTCs in renal carcinoma has not been proven in large scale. However, some studies have reported that specialized markers are found on the surface of circulating cells from the primary tumour. In mice, MICs have been isolated from CTCs using such markers, which have then been transplanted into xenograft model to show whether they give rise to metastasis in different organs. Considering these findings, in this review we have attempted to summarize the studies connected with MICs and their gene expression profiles that are responsible for metastasis in renal cancer.
Collapse
Affiliation(s)
- Mohammed I Khan
- Molecular Oncology Laboratory, Clinic of Oncology, Military Institute of Medicine, ul. Szaserów 128, 04-141 Warsaw, Poland
| | - Anna M Czarnecka
- Molecular Oncology Laboratory, Clinic of Oncology, Military Institute of Medicine, ul. Szaserów 128, 04-141 Warsaw, Poland
| | - Renata Duchnowska
- Molecular Oncology Laboratory, Clinic of Oncology, Military Institute of Medicine, ul. Szaserów 128, 04-141 Warsaw, Poland
| | - Wojciech Kukwa
- Department of Otolaryngology, Czerniakowski Hospital, Medical University of Warsaw, ul. Stepinska 19/25, Warsaw, Poland
| | - Cezary Szczylik
- Molecular Oncology Laboratory, Clinic of Oncology, Military Institute of Medicine, ul. Szaserów 128, 04-141 Warsaw, Poland
| |
Collapse
|
8
|
Ries C. Cytokine functions of TIMP-1. Cell Mol Life Sci 2014; 71:659-72. [PMID: 23982756 PMCID: PMC11113289 DOI: 10.1007/s00018-013-1457-3] [Citation(s) in RCA: 210] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/09/2013] [Accepted: 08/12/2013] [Indexed: 12/13/2022]
Abstract
The tissue inhibitors of metalloproteinases (TIMPs) are well recognized for their role in extracellular matrix remodeling by controlling the activity of matrix metalloproteinases (MMPs). Independent of MMP inhibition, TIMPs act as signaling molecules with cytokine-like activities thereby influencing various biological processes including cell growth, apoptosis, differentiation, angiogenesis, and oncogenesis. Recent studies on TIMP-1's cytokine functions have identified complex regulatory networks involving a specific surface receptor and subsequent signaling pathways including miRNA-mediated posttranscriptional regulation of gene expression that ultimately control the fate and behavior of the cells. The present review summarizes the current knowledge on TIMP-1 as a cytokine modulator of cell functions, outlines recent progress in defining molecular pathways that transmit TIMP-1 signals from the cell periphery into the nucleus, and discusses TIMP-1's role as a cytokine in the pathophysiology of cancer and other human diseases.
Collapse
Affiliation(s)
- Christian Ries
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University of Munich, Pettenkoferstrasse 9b, 80336, Munich, Germany,
| |
Collapse
|
9
|
Recombinant TIMP-1-GPI inhibits growth of fibrosarcoma and enhances tumor sensitivity to doxorubicin. Target Oncol 2013; 9:251-61. [PMID: 23934106 PMCID: PMC4156787 DOI: 10.1007/s11523-013-0294-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 07/26/2013] [Indexed: 12/26/2022]
Abstract
Fibrosarcomas show a high incidence of recurrence and general resistance to apoptosis. Limiting tumor regrowth and increasing their sensitivity to chemotherapy and apoptosis represent key issues in developing more effective treatments of these tumors. Tissue inhibitor of metalloproteinase 1 (TIMP-1) broadly blocks matrix metalloproteinase (MMP) activity and can moderate tumor growth and metastasis. We previously described generation of a recombinant fusion protein linking TIMP-1 to glycosylphophatidylinositol (GPI) anchor (TIMP-1-GPI) that efficiently directs the inhibitor to cell surfaces. In the present report, we examined the effect of TIMP-1-GPI treatment on fibrosarcoma biology. Exogenously applied TIMP-1-GPI efficiently incorporated into surface membranes of human HT1080 fibrosarcoma cells. It inhibited their proliferation, migration, suppressed cancer cell clone formation, and enhanced apoptosis. Doxorubicin, the standard chemotherapeutic drug for fibrosarcoma, was tested alone or in combination with TIMP-1-GPI. In parallel, the influence of treatment on HT1080 side population cells (exhibiting tumor stem cell-like characteristics) was investigated using Hoechst 33342 staining. The sequential combination of TIMP-1-GPI and doxorubicin showed more than additive effects on apoptosis, while TIMP-1-GPI treatment alone effectively decreased “stem-cell like” side population cells of HT1080. TIMP-1-GPI treatment was validated using HT1080 fibrosarcoma murine xenografts. Growing tumors treated with repeated local injections of TIMP-1-GPI showed dramatically inhibited fibrosarcoma growth and reduced angiogenesis. Intraoperative peritumoral application of GPI-anchored TIMP-1 as an adjuvant to surgery may help maintain tumor control by targeting microscopic residual fibrosarcoma cells and increasing their sensitivity to chemotherapy
Collapse
|
10
|
Treatment of dermal fibroblasts with GPI-anchored human TIMP-1 protein moderates processes linked to scar formation. J Invest Dermatol 2012; 133:803-811. [PMID: 23096710 DOI: 10.1038/jid.2012.375] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tissue inhibitors of metalloproteinases exhibit diverse physiological/biological functions including moderation of the proteolytic processing of growth factors and turnover of extracellular matrix. These various biological activities are linked in part to the stoichiometry of tissue inhibitor of metalloprotein/matrix metalloprotein (TIMP/MMP)/surface protein interactions. TIMP-1, a secreted protein, can be detected on the cell surface only through its interaction with surface-bound proteins. Proteins anchored by glycosylphosphatidylinositol (GPI), when purified and added to cells or tissues, are efficiently incorporated into their surface membranes. A GPI anchor was fused to TIMP-1 to focus defined concentrations of the inhibitory protein independently on the surface of primary dermal fibroblast cells. Exogenously added recombinant TIMP-1-GPI effectively inserted into the cell membrane of fibroblasts blocked the secretion of MMPs and markedly altered the stoichiometry of MMP association with the cell surface. TIMP-1-GPI treatment resulted in inhibition of fibroblast-reduced proliferation, and transiently reduced expression of fibrosis-associated genes. These effects were dose dependent. Treated cells also showed a more proapoptotic phenotype based on apoptotic assays and western blot analysis for apoptosis-associated protein expression. GPI-anchored TIMP-1 may represent a more effective version of the protein for use in therapeutic approaches to help control fibrosis and scar formation.
Collapse
|
11
|
Djafarzadeh R, Sauter M, Notohamiprodjo S, Noessner E, Goyal P, Siess W, Wörnle M, Ribeiro A, Himmelein S, Sitter T, Nelson PJ. Recombinant GPI-anchored TIMP-1 stimulates growth and migration of peritoneal mesothelial cells. PLoS One 2012; 7:e33963. [PMID: 22558080 PMCID: PMC3338742 DOI: 10.1371/journal.pone.0033963] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 02/20/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Mesothelial cells are critical in the pathogenesis of post-surgical intraabdominal adhesions as well as in the deterioration of the peritoneal membrane associated with long-term peritoneal dialysis. Mesothelial denudation is a pathophysiolocigally important finding in these processes. Matrix metalloproteinase (MMP) biology underlies aspects of mesothelial homeostasis as well as wound repair. The endogenous tissue inhibitors of metalloproteinases (TIMPs) moderate MMP activity. METHODS AND FINDING By modifying human TIMP-1 through the addition of a glycosylphosphatidylinositol (GPI) anchor, a recombinant protein was generated that efficiently focuses TIMP-1 on the cell surface. Treatment of primary mesothelial cells with TIMP-1-GPI facilitates their mobilization and migration leading to a dramatic increase in the rate of wound experimental closure. Mesothelial cells treated with TIMP-1-GPI showed a dose dependent increase in cell proliferation, reduced secretion of MMP-2, MMP-9, TNF-α and urokinase-type plasminogen activator (uPA), but increased tissue plasminogen activator (t-PA). Treatment resulted in reduced expression and processing of latent TGF-β1. CONCLUSIONS TIMP-1-GPI stimulated rapid and efficient in vitro wound closure. The agent enhanced mesothelial cell proliferation and migration and was bioactive in the nanogram range. The application of TIMP-1-GPI may represent a new approach for limiting or repairing damaged mesothelium.
Collapse
Affiliation(s)
- Roghieh Djafarzadeh
- Arbeitsgruppe Klinische Biochemie, Medizinische Klinik und Poliklinik IV, Universität München, Munich, Germany
| | - Matthias Sauter
- Nephrologie, Medizinische Klinik und Poliklinik IV, Universität München, Munich, Germany
| | - Susan Notohamiprodjo
- Arbeitsgruppe Klinische Biochemie, Medizinische Klinik und Poliklinik IV, Universität München, Munich, Germany
| | - Elfriede Noessner
- Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Pankaj Goyal
- Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten, Klinikum der LMU, München, Germany
| | - Wolfgang Siess
- Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten, Klinikum der LMU, München, Germany
| | - Markus Wörnle
- Nephrologie, Medizinische Klinik und Poliklinik IV, Universität München, Munich, Germany
| | - Andrea Ribeiro
- Nephrologie, Medizinische Klinik und Poliklinik IV, Universität München, Munich, Germany
| | - Susanne Himmelein
- Nephrologie, Medizinische Klinik und Poliklinik IV, Universität München, Munich, Germany
| | - Thomas Sitter
- Nephrologie, Medizinische Klinik und Poliklinik IV, Universität München, Munich, Germany
| | - Peter J. Nelson
- Arbeitsgruppe Klinische Biochemie, Medizinische Klinik und Poliklinik IV, Universität München, Munich, Germany
- * E-mail:
| |
Collapse
|
12
|
Gillet JP, Wang J, Calcagno AM, Green LJ, Varma S, Elstrand MB, Trope CG, Ambudkar SV, Davidson B, Gottesman MM. Clinical relevance of multidrug resistance gene expression in ovarian serous carcinoma effusions. Mol Pharm 2011; 8:2080-8. [PMID: 21761824 PMCID: PMC3224865 DOI: 10.1021/mp200240a] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The presence of tumor cells in effusions within serosal cavities is a clinical manifestation of advanced-stage cancer and is generally associated with poor survival. Identifying molecular targets may help to design efficient treatments to eradicate these aggressive cancer cells and improve patient survival. Using a state-of-the-art TaqMan-based qRT-PCR assay, we investigated the multidrug resistance (MDR) transcriptome of 32 unpaired ovarian serous carcinoma effusion samples obtained at diagnosis or at disease recurrence following chemotherapy. MDR genes were selected a priori based on an extensive curation of the literature published during the last three decades. We found three gene signatures with a statistically significant correlation with overall survival (OS), response to treatment [complete response (CR) vs other], and progression free survival (PFS). The median log-rank p-values for the signatures were 0.023, 0.034, and 0.008, respectively. No correlation was found with residual tumor status after cytoreductive surgery, treatment (with or without chemotherapy) and stage defined according to the International Federation of Gynecology and Obstetrics. Further analyses demonstrated that gene expression alone can effectively predict the survival outcome of women with ovarian serous carcinoma (OS, log-rank p = 0.0000; and PFS, log-rank p = 0.002). Interestingly, the signature for overall survival is the same in patients at first presentation and those who had chemotherapy and relapsed. This pilot study highlights two new gene signatures that may help in optimizing the treatment for ovarian carcinoma patients with effusions.
Collapse
Affiliation(s)
- Jean-Pierre Gillet
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH
| | - Junbai Wang
- Division of Pathology, Norwegian Radium Hospital, Oslo University Hospital, N-0310 Oslo, Norway
| | - Anna Maria Calcagno
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH
| | - Lisa J. Green
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH
| | - Sudhir Varma
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD
| | - Mari Bunkholt Elstrand
- Department of Gynecologic Oncology, Norwegian Radium Hospital, Oslo University Hospital, N-0310 Oslo, Norway
| | - Claes G. Trope
- Department of Gynecologic Oncology, Norwegian Radium Hospital, Oslo University Hospital, N-0310 Oslo, Norway
- The Medical Faculty, University of Oslo, N-0316 Oslo, Norway
| | - Suresh V. Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH
| | - Ben Davidson
- Division of Pathology, Norwegian Radium Hospital, Oslo University Hospital, N-0310 Oslo, Norway
- The Medical Faculty, University of Oslo, N-0316 Oslo, Norway
| | - Michael M. Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH
| |
Collapse
|
13
|
Figel AM, Brech D, Prinz PU, Lettenmeyer UK, Eckl J, Turqueti-Neves A, Mysliwietz J, Anz D, Rieth N, Muenchmeier N, Buchner A, Porubsky S, Siegert SI, Segerer S, Nelson PJ, Noessner E. Human renal cell carcinoma induces a dendritic cell subset that uses T-cell crosstalk for tumor-permissive milieu alterations. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:436-51. [PMID: 21703422 DOI: 10.1016/j.ajpath.2011.03.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 03/17/2011] [Accepted: 03/21/2011] [Indexed: 12/20/2022]
Abstract
Tissue dendritic cells (DCs) may influence the progression of renal cell carcinoma (RCC) by regulating the functional capacity of antitumor effector cells. DCs and their interaction with T cells were analyzed in human RCC and control kidney tissues. The frequency of CD209(+) DCs in RCCs was found to be associated with an unfavorable T(H)1 cell balance in the tissue and advanced tumor stages. The CD209(+) DCs in RCC were unusual because most of them co-expressed macrophage markers (CD14, CD163). The phenotype of these enriched-in-renal-carcinoma DCs (ercDCs) could be reiterated in vitro by carcinoma-secreted factors (CXCL8/IL-8, IL-6, and vascular endothelial growth factor). ErcDCs resembled conventional DCs in costimulatory molecule expression and antigen cross-presentation. They did not suppress cognate cytotoxic T-lymphocyte function and did not cause CD3ζ down-regulation, FOXP3 induction, or T-cell apoptosis in situ or in vitro; thus, they are different from classic myeloid-derived suppressor cells. ErcDCs secreted high levels of metalloproteinase 9 and used T-cell crosstalk to increase tumor-promoting tumor necrosis factor α and reduce chemokines relevant for T(H)1-polarized lymphocyte recruitment. This modulation of the tumor environment exerted by ercDCs suggests an immunologic mechanism by which tumor control can fail without involving cytotoxic T-lymphocyte inhibition. Pharmacologic targeting of the deviated DC differentiation could improve the efficacy of immunotherapy against RCC.
Collapse
Affiliation(s)
- Ainhoa-M Figel
- Institute of Molecular Immunology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Bourquin C, von der Borch P, Zoglmeier C, Anz D, Sandholzer N, Suhartha N, Wurzenberger C, Denzel A, Kammerer R, Zimmermann W, Endres S. Efficient eradication of subcutaneous but not of autochthonous gastric tumors by adoptive T cell transfer in an SV40 T antigen mouse model. THE JOURNAL OF IMMUNOLOGY 2010; 185:2580-8. [PMID: 20644173 DOI: 10.4049/jimmunol.0903231] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In stomach cancer, there is a need for new therapeutic strategies, in particular for the treatment of unresectable tumors and micrometastases. We investigated the efficacy of immunotherapy in an autochthonous model of gastric cancer, the CEA424-SV40 T Ag (TAg) transgenic mice. Treatment efficacy against both the autochthonous tumors and s.c. tumors induced by the derived cell line mGC3 were assessed. In wild-type mice, a dendritic cell vaccine loaded with irradiated tumor cells combined with CpG oligonucleotides induced efficient cytotoxic T cell and memory responses against mGC3 s.c. tumors. In contrast, neither s.c. nor autochthonous tumors responded to vaccination in CEA424-SV40 TAg mice, indicating tolerance to the SV40 TAg. To examine whether tumors in these mice were principally accessible to immunotherapy, splenocytes from immune wild-type mice were adoptively transferred into CEA424-SV40 TAg transgenic mice. Treated mice showed complete regression of the s.c. tumors associated with intratumoral infiltrates of CD8 and CD4 T cells. In contrast, the autochthonous gastric tumors in the same mice were poorly infiltrated and did not regress. Thus, even in the presence of an active anti-tumoral T cell response, autochthonous gastric tumors do not respond to immunotherapy. This is the first comparison of the efficacy of adoptive T cell transfer between transplanted s.c. tumors and autochthonous tumors in the same animals. Our results suggest that in gastric cancer patients, even a strong anti-tumor T cell response will not efficiently penetrate the tumor in the absence of additional therapeutic strategies targeting the tumor microenvironment.
Collapse
Affiliation(s)
- Carole Bourquin
- Division of Clinical Pharmacology, Center of Integrated Protein Science Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Targeting tumor stroma using engineered mesenchymal stem cells reduces the growth of pancreatic carcinoma. Ann Surg 2009; 250:747-53. [PMID: 19826249 DOI: 10.1097/sla.0b013e3181bd62d0] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To analyze the efficacy of engineered mesenchymal stem cell based therapy directed towards pancreatic tumor stroma. SUMMARY BACKGROUND DATA Mesenchymal stem cells (MSC) are actively recruited to tumor stroma where they enhance tumor growth and metastases. Upregulation of chemotactic cytokine (CCL5) by MSCs within the tumor stroma has been shown to play a central role in this process. Murine MSCs were engineered to express reporter genes or therapeutic genes under control of the CCL5 promoter and adoptively transferred into mice with growing pancreatic tumors. The effect on tumor growth and metastases was then evaluated. METHODS MSCs isolated from bone marrow of C57/Bl6 p53 mice were stably transfected with red fluorescent protein (RFP), enhanced green fluorescent protein (eGFP), or herpes simplex virus (HSV) thymidine kinase (Tk) gene driven by the RANTES promoter. MSCs were intravenously applied once per week over 3 weeks to mice carrying an orthotopic, syngeneic pancreatic Panc02 tumor. RESULTS eGFP and RFP signals driven by the CCL5 promoter were detected by fluorescence in treated pancreatic tumor samples. The HSV-Tk therapy group treated intraperitoneal with the prodrug ganciclovir 5 to 7 days after stem cell application lead to a 50% reduction of primary pancreatic tumor growth (P < 0.0003, student t test) and reduced liver metastases (0% vs. 60%). CONCLUSION The active homing of MSCs into primary pancreatic tumor stroma and activation of the CCL5 promoter was verified using eGFP- and RFP-reporter genes. In the presence of ganciclovir, HSV-Tk transfected MSCs led to a significant reduction of primary pancreatic tumor growth and incidence of metastases.
Collapse
|
16
|
Raggi MC, Djafarzadeh R, Muenchmeier N, Hofstetter M, Jahn B, Rieth N, Nelson PJ. Peritumoral administration of GPI-anchored TIMP-1 inhibits colon carcinoma growth in Rag-2 gamma chain-deficient mice. Biol Chem 2009; 390:893-7. [PMID: 19558322 DOI: 10.1515/bc.2009.098] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Exogenous application of recombinant TIMP-1 protein modified by addition of a glycosylphosphatidylinositol (GPI) anchor allows efficient insertion of the fusion protein into cell membranes. This 'cell surface engineering' leads to changes in the proteolytic environment. TIMP-1-GPI shows enhanced as well as novel in vitro biological activities including suppression of proliferation, reduced migration, and inhibition of invasion of the colon carcinoma cell line SW480. Treatment of SW480 tumors implanted in Rag (-/-) common gamma chain (-/-) C57BL/6 mice with peritumorally applied TIMP-1-GPI, control rhTIMP-1 protein, or vehicle shows that TIMP-1-GPI leads to a significant reduction in tumor growth.
Collapse
|
17
|
mRNA expression of matrix metalloproteinases (MMPs) 2 and 9 and tissue inhibitor of matrix metalloproteinases (TIMPs) 1 and 2 in childhood acute lymphoblastic leukemia: potential role of TIMP1 as an adverse prognostic factor. Leuk Res 2009; 34:32-7. [PMID: 19875168 DOI: 10.1016/j.leukres.2009.10.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Revised: 09/24/2009] [Accepted: 10/06/2009] [Indexed: 11/21/2022]
Abstract
This study evaluates the mRNA expression profile of genes TIMP1, TIMP2, MMP2 and MMP9 in diagnostic bone marrow samples from 134 consecutive ALL children by real-time quantitative PCR. A significant association was observed between higher expression levels of MMP9 and low risk group and absence of extramedullary infiltration and higher expression levels of TIMP2 and MMP2 with T-ALL. TIMP1 gene expression values higher than the median were associated with a significantly lower 5-year event free-survival in univariable (P=0.04) and multivariable analysis (P=0.01). Our data address new information in the complex interaction of the migration/adhesion genes and childhood ALL.
Collapse
|
18
|
Djafarzadeh R, Milani V, Rieth N, von Luettichau I, Skrablin PS, Hofstetter M, Noessner E, Nelson PJ. TIMP-1-GPI in combination with hyperthermic treatment of melanoma increases sensitivity to FAS-mediated apoptosis. Cancer Immunol Immunother 2009; 58:361-71. [PMID: 18618109 PMCID: PMC11030769 DOI: 10.1007/s00262-008-0559-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Accepted: 06/27/2008] [Indexed: 02/02/2023]
Abstract
Resistance to apoptosis is a prominent feature of malignant melanoma. Hyperthermic therapy can be an effective adjuvant treatment for some tumors including melanoma. We developed a fusion protein based on the tissue inhibitor of matrix metalloproteinase-1 linked to a glycosylphosphatidylinositol anchor (TIMP-1-GPI). The TIMP-1-GPI-fusion protein shows unique properties. Exogenous administration of TIMP-1-GPI can result in transient morphological changes to treated cells including modulation of proliferation and decreased resistance to apoptosis. The effect of TIMP-1-GPI on the biology of melanoma in the context of a defined hyperthermic dose was evaluated in vitro. Clonogenic assays were used to measure cell survival. Gelatinase zymography determined secretion of MMP-2 and MMP-9. Monoclonal antibody against FAS/CD95 was applied to induce apoptosis. The expression of pro- and anti-apoptotic proteins and the secretion of immunoregulatory cytokines were then evaluated using Western blot and ELISA. TIMP-1-GPI combined with a sub-lethal hyperthermic treatment (41.8 degrees C for 2 h) suppressed tumor cell growth capacity as measured by clonogenic assay. The co-treatment also significantly suppressed tumor cell proliferation, enhanced FAS receptor surface expression increased tumor cell susceptibility to FAS-mediated killing. The increased sensitivity to FAS-induced apoptosis was linked to alterations in the apoptotic mediators Bcl-2, Bax, Bcl-XL and Apaf-1. The agent works in concert with sub-lethal hyperthermic treatment to render melanoma cells sensitive to FAS killing. The targeted delivery of TIMP-1-GPI to tumor environments in the context of regional hyperthermic therapy could be optimized through the use of thermosensitive liposomes.
Collapse
Affiliation(s)
- Roghieh Djafarzadeh
- Medizinische Poliklinik, Ludwig-Maximilians-Universität München, Schillerstrasse 42, 80336 Munich, Germany
| | - Valeria Milani
- Medical Clinic III, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Nicole Rieth
- Medizinische Poliklinik, Ludwig-Maximilians-Universität München, Schillerstrasse 42, 80336 Munich, Germany
| | - Irene von Luettichau
- Medizinische Poliklinik, Ludwig-Maximilians-Universität München, Schillerstrasse 42, 80336 Munich, Germany
- Department of Paediatrics, Technical University, Munich, Germany
| | - Petra S. Skrablin
- Helmholz Zentrum Munich-Institute of Molecular Immunology, Munich, Germany
| | - Monika Hofstetter
- Medizinische Poliklinik, Ludwig-Maximilians-Universität München, Schillerstrasse 42, 80336 Munich, Germany
| | - Elfriede Noessner
- Helmholz Zentrum Munich-Institute of Molecular Immunology, Munich, Germany
| | - Peter J. Nelson
- Medizinische Poliklinik, Ludwig-Maximilians-Universität München, Schillerstrasse 42, 80336 Munich, Germany
| |
Collapse
|
19
|
Schuster IG, Busch DH, Eppinger E, Kremmer E, Milosevic S, Hennard C, Kuttler C, Ellwart JW, Frankenberger B, Nössner E, Salat C, Bogner C, Borkhardt A, Kolb HJ, Krackhardt AM. Allorestricted T cells with specificity for the FMNL1-derived peptide PP2 have potent antitumor activity against hematologic and other malignancies. Blood 2007; 110:2931-9. [PMID: 17626842 DOI: 10.1182/blood-2006-11-058750] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
AbstractCell-based immunotherapy in settings of allogeneic stem cell transplantation or donor leukocyte infusion has curative potential, especially in hematologic malignancies. However, this approach is severely restricted due to graft-versus-host disease (GvHD). This limitation may be overcome if target antigens are molecularly defined and effector cells are specifically selected. We chose formin-related protein in leukocytes 1 (FMNL1) as a target antigen after intensive investigation of its expression profile at the mRNA and protein levels. Here, we confirm restricted expression in peripheral blood mononuclear cells (PBMCs) from healthy donors but also observe overexpression in different leukemias and aberrant expression in transformed cell lines derived from solid tumors. We isolated allorestricted T-cell clones expressing a single defined TCR recognizing a particular HLA-A2–presented peptide derived from FMNL1. This T-cell clone showed potent antitumor activity against lymphoma and renal cell carcinoma cell lines, Epstein-Barr virus (EBV)–transformed B cells, and primary tumor samples derived from patients with chronic lymphocytic leukemia (CLL), whereas nontransformed cells with the exception of activated B cells were only marginally recognized. Allorestricted TCRs with specificity for naturally presented FMNL1-derived epitopes may represent promising reagents for the development of adoptive therapies in lymphoma and other malignant diseases.
Collapse
MESH Headings
- Antigens, Neoplasm/immunology
- Blotting, Western
- Bone Marrow/metabolism
- Cell Line, Tumor
- Clone Cells
- Cytoskeletal Proteins/immunology
- Cytoskeletal Proteins/metabolism
- Cytotoxicity, Immunologic
- Enzyme-Linked Immunosorbent Assay
- Epitopes, T-Lymphocyte/immunology
- Formins
- HLA-A Antigens
- Hematologic Neoplasms/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Leukocytes, Mononuclear/metabolism
- Peptides/immunology
- Receptors, Antigen, T-Cell/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes, Cytotoxic/immunology
- Thymus Gland/metabolism
Collapse
Affiliation(s)
- Ingrid G Schuster
- Institute of Molecular Immunology, Forschungszeutrum für Umwelt und Gesundheit (GSF)-National Research Center for Environment and Health, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Frankenberger B, Noessner E, Schendel DJ. Immune suppression in renal cell carcinoma. Semin Cancer Biol 2007; 17:330-43. [PMID: 17656104 DOI: 10.1016/j.semcancer.2007.06.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Revised: 04/10/2007] [Accepted: 06/07/2007] [Indexed: 12/13/2022]
Abstract
The clear evidence that tumor-infiltrating lymphocytes with anti-tumor activity exist in situ raises the question why renal cell carcinomas (RCCs) progress in vivo. A complex array of factors and pathways has been identified that impinges on innate and adaptive effector cells thereby inhibiting their activity against RCCs. The current picture of suppressive mechanisms that contribute to the failure of the immune system to control RCCs is reviewed here. Understanding these complex host-tumor interactions has broad implications for successful application of cytokine therapy and other forms of immunotherapy for RCC.
Collapse
Affiliation(s)
- Bernhard Frankenberger
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Marchioninistrasse 25, 81377 Munich, Germany
| | | | | |
Collapse
|
21
|
Jiang H, Zhu H, Chen X, Peng Y, Wang J, Liu F, Shi S, Fu B, Lu Y, Hong Q, Feng Z, Hou K, Sun X, Cai G, Zhang X, Xie Y. TIMP-1 transgenic mice recover from diabetes induced by multiple low-dose streptozotocin. Diabetes 2007; 56:49-56. [PMID: 17192464 DOI: 10.2337/db06-0710] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Type 1 diabetes results from autoimmune destruction of the insulin-producing beta-cells of pancreatic islets, of which the capacity for self-replication in the adult is too limited to restore following extensive tissue injury. Tissue inhibitor of metalloproteinase (TIMP)-1 inhibits matrix metalloproteinase activity and regulates proliferation and apoptosis of a variety of cells types, depending on the context. Here, we show that overexpression of human TIMP-1 in pancreatic beta-cells of transgenic mice counteracts the cytotoxicity and insulitis induced by multiple low-dose streptozotocin (MLDS). Nontransgenic mice developed severe hyperglycemia, hypoinsulinemia, and insulitis 2 weeks after streptozotocin administration and died within 17 weeks. However, MLDS-treated transgenic mice gradually normalized the metabolic parameters and survived. beta-Cell mass increased in parallel as a result of enhancement of beta-cell replication. Thus, our results have demonstrated for the first time that overexpression of TIMP-1 in beta-cells enhances the replication of pancreatic islets beta-cells and counteracts type 1 diabetes, indicating that the TIMP-1 gene may be a potential target to prevent, or even reverse, type 1 diabetes.
Collapse
Affiliation(s)
- Hongwei Jiang
- Department of Nephrology, Kidney Center and Key Lab of People's Liberation Army, General Hospital of PLA, Fuxing Road 28, Beijing 100853, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Geiger C, Regn S, Weinzierl A, Noessner E, Schendel DJ. A generic RNA-pulsed dendritic cell vaccine strategy for renal cell carcinoma. J Transl Med 2005; 3:29. [PMID: 16045799 PMCID: PMC1188079 DOI: 10.1186/1479-5876-3-29] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2005] [Accepted: 07/26/2005] [Indexed: 11/18/2022] Open
Abstract
We present a generic dendritic cell (DC) vaccine strategy for patients with renal cell carcinoma (RCC) based on the use of RNA as a source of multiplex tumor-associated antigens (TAAs). Instead of preparing RNA from tumor tissue of each individual RCC patient, we propose to substitute RNA prepared from a well characterized highly immunogenic RCC cell line (RCC-26 tumor cells) as a generic source of TAAs for loading of DCs. We demonstrate here that efficient RNA transfer can be achieved using lipofection of immature DCs, which are subsequently matured with a cytokine cocktail to express high levels of MHC and costimulatory molecules as well as the chemokine receptor CCR7. Neither RNA itself nor the lipid component impacted on the phenotype or the cytokine secretion of mature DCs. Following RNA loading, DCs derived from HLA-A2-positive donors were able to activate effector-memory cytotoxic T lymphocytes (CTLs) specific for a TAA ligand expressed by the RCC-26 cell line. CTL responses to RNA-loaded DCs reached levels comparable to those stimulated directly by the RCC-26 tumor cells. Furthermore, DCs expressing tumor cell RNA primed naïve T cells, yielding T cell lines with cytotoxicity and cytokine secretion after contact with RCC tumor cells. RCC-26 cell lines are available as good manufacturing practice (GMP)-certified reagents enabling this source of RNA to be easily standardized and adapted for clinical testing. In addition, well defined immune monitoring tools, including the use of RNA expressing B cell lines, are available. Thus, this DC vaccine strategy can be directly compared with an ongoing gene therapy trial using genetically-engineered variants of the RCC-26 cell line as vaccines for RCC patients with metastatic disease.
Collapse
Affiliation(s)
- Christiane Geiger
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Munich, Germany
| | - Sybille Regn
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Munich, Germany
| | - Andreas Weinzierl
- Institute of Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Elfriede Noessner
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Munich, Germany
| | - Dolores J Schendel
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Munich, Germany
| |
Collapse
|