51
|
Shevtsov M, Huile G, Multhoff G. Membrane heat shock protein 70: a theranostic target for cancer therapy. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0526. [PMID: 29203711 PMCID: PMC5717526 DOI: 10.1098/rstb.2016.0526] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2017] [Indexed: 12/19/2022] Open
Abstract
Members of the 70 kDa stress protein family are found in nearly all subcellular compartments of nucleated cells where they fulfil a number of chaperoning functions. Heat shock protein 70 (HSP70), also termed HSPA1A, the major stress-inducible member of this family is overexpressed in a large variety of different tumour types. Apart from its intracellular localization, a tumour-selective HSP70 membrane expression has been determined. A membrane HSP70–positive tumour phenotype is associated with aggressiveness and therapy resistance, but also serves as a recognition structure for targeted therapies. Furthermore, membrane-bound and extracellularly residing HSP70 derived from tumour cells play pivotal roles in eliciting anti-tumour immune responses. Herein, we want to shed light on the multiplicity of different activities of HSP70, depending on its intracellular, membrane and extracellular localization with the goal to use membrane HSP70 as a target for novel therapies including nanoparticle-based approaches for the treatment of cancer. This article is part of the theme issue ‘Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective’.
Collapse
Affiliation(s)
- Maxim Shevtsov
- Klinikum rechts der Isar, Department of Radiation Oncology, Technische Universität München, Ismaninger Strasse 22, Munich 81675, Germany.,Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Avenue, 4, St Petersburg 194064, Russia
| | - Gao Huile
- West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Gabriele Multhoff
- Klinikum rechts der Isar, Department of Radiation Oncology, Technische Universität München, Ismaninger Strasse 22, Munich 81675, Germany
| |
Collapse
|
52
|
Tag7 (PGLYRP1) Can Induce an Emergence of the CD3+CD4+CD25+CD127+ Cells with Antitumor Activity. J Immunol Res 2018; 2018:4501273. [PMID: 29850628 PMCID: PMC5925135 DOI: 10.1155/2018/4501273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/11/2018] [Accepted: 02/11/2018] [Indexed: 11/17/2022] Open
Abstract
We have shown that in the human peripheral blood cells, the innate immunity protein Tag7 can activate a subpopulation of CD3+CD4+CD25+ cells, which have antitumor activity. These cells can induce lysis of HLA-negative tumor cell lines. The Hsp70 stress molecule on the surface of the tumor cells is used as a recognition target, while the Tag7 protein on the lymphocyte membrane acts as a receptor for Hsp70. We have also demonstrated that this subpopulation of the CD4+CD25+ cells is CD127 positive and hence is not the Treg cells. Our data suggest that this subpopulation of cells is identical to the CD4+CD25+ lymphocytes, which are activated in the leukocyte pool by the IL-2 cytokine.
Collapse
|
53
|
Stangl S, Tontcheva N, Sievert W, Shevtsov M, Niu M, Schmid TE, Pigorsch S, Combs SE, Haller B, Balermpas P, Rödel F, Rödel C, Fokas E, Krause M, Linge A, Lohaus F, Baumann M, Tinhofer I, Budach V, Stuschke M, Grosu AL, Abdollahi A, Debus J, Belka C, Maihöfer C, Mönnich D, Zips D, Multhoff G. Heat shock protein 70 and tumor-infiltrating NK cells as prognostic indicators for patients with squamous cell carcinoma of the head and neck after radiochemotherapy: A multicentre retrospective study of the German Cancer Consortium Radiation Oncology Group (DKTK-ROG). Int J Cancer 2017; 142:1911-1925. [PMID: 29235112 PMCID: PMC5873418 DOI: 10.1002/ijc.31213] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/03/2017] [Accepted: 11/28/2017] [Indexed: 12/28/2022]
Abstract
Tumor cells frequently overexpress heat shock protein 70 (Hsp70) and present it on their cell surface, where it can be recognized by pre-activated NK cells. In our retrospective study the expression of Hsp70 was determined in relation to tumor-infiltrating CD56+ NK cells in formalin-fixed paraffin embedded (FFPE) tumor specimens of patients with SCCHN (N = 145) as potential indicators for survival and disease recurrence. All patients received radical surgery and postoperative cisplatin-based radiochemotherapy (RCT). In general, Hsp70 expression was stronger, but with variable intensities, in tumor compared to normal tissues. Patients with high Hsp70 expressing tumors (scores 3-4) showed significantly decreased overall survival (OS; p = 0.008), local progression-free survival (LPFS; p = 0.034) and distant metastases-free survival (DMFS; p = 0.044), compared to those with low Hsp70 expression (scores 0-2), which remained significant after adjustment for relevant prognostic variables. The adverse prognostic value of a high Hsp70 expression for OS was also observed in patient cohorts with p16- (p = 0.001), p53- (p = 0.0003) and HPV16 DNA-negative (p = 0.001) tumors. The absence or low numbers of tumor-infiltrating CD56+ NK cells also correlated with significantly decreased OS (p = 0.0001), LPFS (p = 0.0009) and DMFS (p = 0.0001). A high Hsp70 expression and low numbers of tumor-infiltrating NK cells have the highest negative predictive value (p = 0.00004). In summary, a strong Hsp70 expression and low numbers of tumor-infiltrating NK cells correlate with unfavorable outcome following surgery and RCT in patients with SCCHN, and thus serve as negative prognostic markers.
Collapse
Affiliation(s)
- Stefan Stangl
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| | | | - Wolfgang Sievert
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| | - Maxim Shevtsov
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| | - Minli Niu
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany
| | - Thomas E Schmid
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| | - Steffi Pigorsch
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| | - Bernhard Haller
- Institute of Medical Informatics, Statistics and Epidemiology, Technische Universität München (TUM), Munich, Germany
| | - Panagiotis Balermpas
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Frankfurt, Frankfurt, Germany
| | - Franz Rödel
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Frankfurt, Frankfurt, Germany
| | - Claus Rödel
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Frankfurt, Frankfurt, Germany
| | - Emmanouil Fokas
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Frankfurt, Frankfurt, Germany
| | - Mechthild Krause
- OncoRay - National Centre for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Dresden, Germany.,Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology, Berlin, Germany
| | - Annett Linge
- OncoRay - National Centre for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Dresden, Germany
| | - Fabian Lohaus
- OncoRay - National Centre for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Dresden, Germany
| | - Michael Baumann
- OncoRay - National Centre for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Dresden, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Heidelberg, Germany
| | - Inge Tinhofer
- Department of Radiooncology and Radiotherapy, Charite University Hospital Berlin, Berlin, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Berlin, Germany
| | - Volker Budach
- Department of Radiooncology and Radiotherapy, Charite University Hospital Berlin, Berlin, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Berlin, Germany
| | - Martin Stuschke
- Department of Radiotherapy, Medical Faculty, University of Duisburg-Essen, Essen, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Essen, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Faculty, Medical Centre, University of Freiburg, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Freiburg, Germany
| | - Amir Abdollahi
- German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Heidelberg, Germany
| | - Jürgen Debus
- German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg Ion Therapy Centre (HIT), Heidelberg Institute of Radiation Oncology (HIRO), University of Heidelberg Medical School, Heidelberg, Germany.,National Centre for Radiation Research Oncology (NCRO), University of Heidelberg Medical School, Heidelberg, Germany
| | - Claus Belka
- German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany.,National Centre for Radiation Research Oncology (NCRO), University of Heidelberg Medical School, Heidelberg, Germany.,Department of Radiation Oncology, University Hospital LMU Munich, Munich, Germany.,Clinical Cooperation Group (CCG) Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum, Munich, Germany
| | - Cornelius Maihöfer
- German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany.,National Centre for Radiation Research Oncology (NCRO), University of Heidelberg Medical School, Heidelberg, Germany.,Department of Radiation Oncology, University Hospital LMU Munich, Munich, Germany.,Clinical Cooperation Group (CCG) Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum, Munich, Germany
| | - David Mönnich
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany.,DKTK Consortium Tübingen, Tübingen, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany.,DKTK Consortium Tübingen, Tübingen, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Tübingen, Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München (TUM), Munich, Germany.,Department of Radiation Sciences (DRS), Helmholtz Zentrum Munich (HMGU), Institute of Innovative Radiotherapy (iRT), Munich, Germany.,German Cancer Research Centre (DKFZ), Heidelberg and German Research Consortium (DKTK), Munich, Germany
| |
Collapse
|
54
|
Friedrich L, Kornberger P, Mendler CT, Multhoff G, Schwaiger M, Skerra A. Selection of an Anticalin® against the membrane form of Hsp70 via bacterial surface display and its theranostic application in tumour models. Biol Chem 2017; 399:235-252. [DOI: 10.1515/hsz-2017-0207] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/23/2017] [Indexed: 01/08/2023]
Abstract
Abstract
We describe the selection of Anticalins against a common tumour surface antigen, human Hsp70, using functional display on live Escherichia coli cells as fusion with a truncated EspP autotransporter. While found intracellularly in normal cells, Hsp70 is frequently exposed in a membrane-bound state on the surface of tumour cells and, even more pronounced, in metastases or after radiochemotherapy. Employing a recombinant Hsp70 fragment comprising residues 383-548 as the target, Anticalins were selected from a naïve bacterial library. The Anticalin with the highest affinity (K
D=13 nm), as determined towards recombinant full-length Hsp70 by real-time surface plasmon resonance analysis, was improved to K
D=510 pm by doped random mutagenesis and another cycle of E. coli surface display, followed by rational combination of mutations. This Anticalin, which recognises a linear peptide epitope located in the interdomain linker of Hsp70, was demonstrated to specifically bind Hsp70 in its membrane-associated form in immunofluorescence microscopy and via flow cytometry using the FaDu cell line, which is positive for surface Hsp70. The radiolabelled and PASylated Anticalin revealed specific tumour accumulation in xenograft mice using positron emission tomography (PET) imaging. Furthermore, after enzymatic coupling to the protein toxin gelonin, the Anticalin showed potent cytotoxicity on FaDu cells in vitro.
Collapse
Affiliation(s)
- Lars Friedrich
- Munich Center for Integrated Protein Science, CIPS-M, and Lehrstuhl für Biologische Chemie , Technische Universität München , D-85354 Freising (Weihenstephan) , Germany
| | - Petra Kornberger
- Munich Center for Integrated Protein Science, CIPS-M, and Lehrstuhl für Biologische Chemie , Technische Universität München , D-85354 Freising (Weihenstephan) , Germany
| | - Claudia T. Mendler
- Munich Center for Integrated Protein Science, CIPS-M, and Lehrstuhl für Biologische Chemie , Technische Universität München , D-85354 Freising (Weihenstephan) , Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology , Klinikum rechts der Isar, Technische Universität München , D-81675 München , Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar , Technische Universität München , D-81675 München , Germany
| | - Arne Skerra
- Munich Center for Integrated Protein Science, CIPS-M, and Lehrstuhl für Biologische Chemie , Technische Universität München , D-85354 Freising (Weihenstephan) , Germany
| |
Collapse
|
55
|
Zeni O, Simkó M, Scarfi MR, Mattsson MO. Cellular Response to ELF-MF and Heat: Evidence for a Common Involvement of Heat Shock Proteins? Front Public Health 2017; 5:280. [PMID: 29094036 PMCID: PMC5651525 DOI: 10.3389/fpubh.2017.00280] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 10/02/2017] [Indexed: 11/13/2022] Open
Abstract
It has been shown that magnetic fields in the extremely low frequency range (ELF-MF) can act as a stressor in various in vivo or in vitro systems, at flux density levels below those inducing excitation of nerve and muscle cells, which are setting the limits used by most generally accepted exposure guidelines, such as the ones published by the International Commission on Non-Ionizing Radiation Protection. In response to a variety of physiological and environmental factors, including heat, cells activate an ancient signaling pathway leading to the transient expression of heat shock proteins (HSPs), which exhibit sophisticated protection mechanisms. A number of studies suggest that also ELF-MF exposure can activate the cellular stress response and cause increased HSPs expression, both on the mRNA and the protein levels. In this review, we provide some of the presently available data on cellular responses, especially regarding HSP expression, due to single and combined exposure to ELF-MF and heat, with the aim to compare the induced effects and to detect possible common modes of action. Some evidence suggest that MF and heat can act as costressors inducing a kind of thermotolerance in cell cultures and in organisms. The MF exposure might produce a potentiated or synergistic biological response such as an increase in HSPs expression, in combination with a well-defined stress, and in turn exert beneficial effects during certain circumstances.
Collapse
Affiliation(s)
- Olga Zeni
- Institute for Electromagnetic Sensing of the Environment (IREA), National Research Council, Naples, Italy
| | | | - Maria Rosaria Scarfi
- Institute for Electromagnetic Sensing of the Environment (IREA), National Research Council, Naples, Italy
| | - Mats-Olof Mattsson
- AIT Austrian Institute of Technology, Center for Energy, Environmental Resources and Technologies, Tulln, Austria
| |
Collapse
|
56
|
Thorsteinsdottir J, Stangl S, Fu P, Guo K, Albrecht V, Eigenbrod S, Erl J, Gehrmann M, Tonn JC, Multhoff G, Schichor C. Overexpression of cytosolic, plasma membrane bound and extracellular heat shock protein 70 (Hsp70) in primary glioblastomas. J Neurooncol 2017; 135:443-452. [PMID: 28849427 DOI: 10.1007/s11060-017-2600-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 08/19/2017] [Indexed: 01/16/2023]
Abstract
A unique feature in several non-CNS-tumors is the overexpression of heat shock protein 70 (Hsp70, HSPA1A) in the cytosol, but also its unusual plasma membrane expression and release. Although in gliomas, cytosolic Hsp70 levels are not associated with histological grading, the role of membrane bound and released Hsp70 is still completely unknown. Membrane bound as well as cytosolic Hsp70 can be detected in viable tumor cells with the monoclonal antibody (mAb) cmHsp70.1. Herein, we analysed membrane bound Hsp70 levels in primary and secondary gliomas of different grades and on isolated glioma subpopulations (endothelial cells, CD133-positive cells, primary cultures) by immunohistochemistry and flow cytometry using cmHsp70.1 mAb. Extracellular Hsp70 was determined by a commercial Hsp70 sandwich ELISA (R&D) in plasma samples of glioblastoma patients and healthy volunteers. We found an overexpression of Hsp70 in primary glioblastomas compared to low-grade, anaplastic, or secondary gliomas as determined by immunohistochemistry. Especially in flow cytometry, a strong plasma membrane Hsp70 expression was only observed in primary but not secondary glioblastomas. Within the heterogeneous tumor mass, CD133-positive tumor-initiating and primary glioblastoma cells showed a high membrane Hsp70 expression density, whereas endothelial cells, isolated from glioblastoma tissues only showed a weak staining pattern. Also in plasma samples, secreted Hsp70 protein was significantly increased in patients harbouring primary glioblastomas compared to those with secondary and low grade glioblastomas. Taken together, we show for the first time that cytosolic, membrane bound and extracellular Hsp70 is uniquely overexpressed in primary glioblastomas.
Collapse
Affiliation(s)
- Jun Thorsteinsdottir
- Tumorbiological Laboratory, Department of Neurosurgery, Ludwig-Maximilians-University, Klinikum Grosshadern, Marchioninistr. 15, 81377, Munich, Germany.
| | - Stefan Stangl
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technical University (TU) Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Peng Fu
- Tumorbiological Laboratory, Department of Neurosurgery, Ludwig-Maximilians-University, Klinikum Grosshadern, Marchioninistr. 15, 81377, Munich, Germany.,Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue No. 1277, Wuhan, 430022, China
| | - Ketai Guo
- Tumorbiological Laboratory, Department of Neurosurgery, Ludwig-Maximilians-University, Klinikum Grosshadern, Marchioninistr. 15, 81377, Munich, Germany
| | - Valerie Albrecht
- Department of Radiation Oncology, Ludwig-Maximilians-University, Klinikum Grosshadern, Marchioninistr. 15, 81377, Munich, Germany
| | - Sabina Eigenbrod
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Feodor-Lynen-Str. 20, 81377, Munich, Germany
| | - Janina Erl
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technical University (TU) Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Mathias Gehrmann
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technical University (TU) Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Jörg-Christian Tonn
- Tumorbiological Laboratory, Department of Neurosurgery, Ludwig-Maximilians-University, Klinikum Grosshadern, Marchioninistr. 15, 81377, Munich, Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technical University (TU) Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Christian Schichor
- Tumorbiological Laboratory, Department of Neurosurgery, Ludwig-Maximilians-University, Klinikum Grosshadern, Marchioninistr. 15, 81377, Munich, Germany
| |
Collapse
|
57
|
Barreca MM, Spinello W, Cavalieri V, Turturici G, Sconzo G, Kaur P, Tinnirello R, Asea AAA, Geraci F. Extracellular Hsp70 Enhances Mesoangioblast Migration via an Autocrine Signaling Pathway. J Cell Physiol 2017; 232:1845-1861. [PMID: 27925208 DOI: 10.1002/jcp.25722] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 11/29/2016] [Indexed: 12/20/2022]
Abstract
Mouse mesoangioblasts are vessel-associated progenitor stem cells endowed with the ability of multipotent mesoderm differentiation. Therefore, they represent a promising tool in the regeneration of injured tissues. Several studies have demonstrated that homing of mesoangioblasts into blood and injured tissues are mainly controlled by cytokines/chemokines and other inflammatory factors. However, little is known about the molecular mechanisms regulating their ability to traverse the extracellular matrix (ECM). Here, we demonstrate that membrane vesicles released by mesoangioblasts contain Hsp70, and that the released Hsp70 is able to interact by an autocrine mechanism with Toll-like receptor 4 (TLR4) and CD91 to stimulate migration. We further demonstrate that Hsp70 has a positive role in regulating matrix metalloproteinase 2 (MMP2) and MMP9 expression and that MMP2 has a more pronounced effect on cell migration, as compared to MMP9. In addition, the analysis of the intracellular pathways implicated in Hsp70 regulated signal transduction showed the involvement of both PI3K/AKT and NF-κB. Taken together, our findings present a paradigm shift in our understanding of the molecular mechanisms that regulate mesoangioblast stem cells ability to traverse the extracellular matrix (ECM). J. Cell. Physiol. 232: 1845-1861, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Maria M Barreca
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Walter Spinello
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Vincenzo Cavalieri
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Giuseppina Turturici
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Gabriella Sconzo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Punit Kaur
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia
| | - Rosaria Tinnirello
- Biomedicine and Molecular Immunology Institute, National Center of Research, Palermo, Italy
| | - Alexzander A A Asea
- Department of Neurology and the Deanship for Scientific Research, University of Dammam, Dammam, Saudi Arabia
| | - Fabiana Geraci
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy.,Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
| |
Collapse
|
58
|
Fitter S, Gronthos S, Ooi SS, Zannettino AC. The Mesenchymal Precursor Cell Marker Antibody STRO-1 Binds to Cell Surface Heat Shock Cognate 70. Stem Cells 2017; 35:940-951. [DOI: 10.1002/stem.2560] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/25/2016] [Accepted: 12/02/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Stephen Fitter
- Myeloma Research Laboratory, Faculty of Health and Medical Science, Adelaide Medical School
- Cancer Theme, South Australian Health and Medical Research Institute; Adelaide South Australia Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide; Adelaide South Australia Australia
- Cancer Theme, South Australian Health and Medical Research Institute; Adelaide South Australia Australia
| | - Soo Siang Ooi
- Myeloma Research Laboratory, Faculty of Health and Medical Science, Adelaide Medical School
- Cancer Theme, South Australian Health and Medical Research Institute; Adelaide South Australia Australia
| | - Andrew C.W. Zannettino
- Myeloma Research Laboratory, Faculty of Health and Medical Science, Adelaide Medical School
- Cancer Theme, South Australian Health and Medical Research Institute; Adelaide South Australia Australia
| |
Collapse
|
59
|
Juric MK, Shevtsov M, Mozes P, Ogonek J, Crossland RE, Dickinson AM, Greinix HT, Holler E, Weissinger EM, Multhoff G. B-Cell-Based and Soluble Biomarkers in Body Liquids for Predicting Acute/Chronic Graft-versus-Host Disease after Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2017; 7:660. [PMID: 28138325 PMCID: PMC5238459 DOI: 10.3389/fimmu.2016.00660] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/16/2016] [Indexed: 02/02/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the main curative therapy for hematological malignancy such as leukemias, lymphomas, or multiple myelomas and some other hematological disorders. In this therapy, cure of hematological diseases relies on graft-versus-malignancy effects by allogenic immune cells. However, severe posttransplant treatment-associated complications such as acute graft-versus-host disease (aGvHD) and chronic graft-versus-host disease (cGvHD) limit this approach. Most research into GvHD has concentrated on the aGvHD, while the more complex and multifaceted chronic form has been largely poorly investigated. cGvHD is a multi-organ autoimmune disorder and is the major cause of non-relapse morbidity and mortality following allo-HSCT, occurring in about 50% of patients, or 13,000–15,000 patients per year worldwide. Therefore, there is a high medical need for an early prediction of these therapy-associated toxicities. Biomarkers have gained importance over the last decade in diagnosis, in prognosis, and in prediction of pending diseases or side effects. Biomarkers can be cells, factors isolated from target tissues, or soluble factors that can be detected in body fluids. In this review, we aim to summarize some of the recent developments of biomarkers in the field of allo-HSCT. We will focus on cell-based biomarkers (B-cell subsets) for cGvHD and soluble factors including microRNA (miRNA), which are excreted into serum/plasma and urine. We also discuss the potential role of cytosolic and extracellular 70 kDa heat shock proteins (HSP70) as potential biomarkers for aGvHD and their role in preclinical models. Proteomic biomarkers in the blood have been used as predictors of treatment responses in patients with aGvHD for many years. More recently, miRNAs have been found to serve as a biomarker to diagnose aGvHD in the plasma. Another development relates to urine-based biomarkers that are usually detected by capillary electrophoresis and mass spectrometry. These biomarkers have the potential to predict the development of severe aGvHD (grades III–IV), overall mortality, and the pending development of cGvHD in patients posttransplant.
Collapse
Affiliation(s)
- Mateja Kralj Juric
- Department of Internal Medicine I, BMT, Medical University of Vienna , Vienna , Austria
| | - Maxim Shevtsov
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| | - Petra Mozes
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| | - Justyna Ogonek
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Transplantation Biology, Hannover Medical School , Hannover , Germany
| | - Rachel E Crossland
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | - Anne M Dickinson
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | | | - Ernst Holler
- Department of Internal Medicine III, University Hospital of Regensburg , Regensburg , Germany
| | - Eva M Weissinger
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Transplantation Biology, Hannover Medical School , Hannover , Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| |
Collapse
|
60
|
Batulan Z, Pulakazhi Venu VK, Li Y, Koumbadinga G, Alvarez-Olmedo DG, Shi C, O'Brien ER. Extracellular Release and Signaling by Heat Shock Protein 27: Role in Modifying Vascular Inflammation. Front Immunol 2016; 7:285. [PMID: 27507972 PMCID: PMC4960997 DOI: 10.3389/fimmu.2016.00285] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/14/2016] [Indexed: 12/31/2022] Open
Abstract
Heat shock protein 27 (HSP27) is traditionally viewed as an intracellular chaperone protein with anti-apoptotic properties. However, recent data indicate that a number of heat shock proteins, including HSP27, are also found in the extracellular space where they may signal via membrane receptors to alter gene transcription and cellular function. Therefore, there is increasing interest in better understanding how HSP27 is released from cells, its levels and composition in the extracellular space, and the cognate cell membrane receptors involved in effecting cell signaling. In this paper, the knowledge to date, as well as some emerging paradigms about the extracellular function of HSP27 is presented. Of particular interest is the role of HSP27 in attenuating atherogenesis by modifying lipid uptake and inflammation in the plaque. Moreover, the abundance of HSP27 in serum is an emerging new biomarker for ischemic events. Finally, HSP27 replacement therapy may represent a novel therapeutic opportunity for chronic inflammatory disorders, such as atherosclerosis.
Collapse
Affiliation(s)
- Zarah Batulan
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Vivek Krishna Pulakazhi Venu
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Yumei Li
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Geremy Koumbadinga
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Daiana Gisela Alvarez-Olmedo
- Oncology Laboratory, Institute for Experimental Medicine and Biology of Cuyo (IMBECU), CCT CONICET , Mendoza , Argentina
| | - Chunhua Shi
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Edward R O'Brien
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| |
Collapse
|
61
|
Lopez V, Cauvi DM, Arispe N, De Maio A. Bacterial Hsp70 (DnaK) and mammalian Hsp70 interact differently with lipid membranes. Cell Stress Chaperones 2016; 21:609-16. [PMID: 27075190 PMCID: PMC4907991 DOI: 10.1007/s12192-016-0685-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/16/2016] [Accepted: 03/17/2016] [Indexed: 10/22/2022] Open
Abstract
The cellular response to stress is orchestrated by the expression of a family of proteins termed heat shock proteins (hsp) that are involved in the stabilization of basic cellular processes to preserve cell viability and homeostasis. The bulk of hsp function occurs within the cytosol and subcellular compartments. However, some hsp have also been found outside cells released by an active mechanism independent of cell death. Extracellular hsp act as signaling molecules directed at activating a systemic response to stress. The export of hsp requires the translocation from the cytosol into the extracellular milieu across the plasma membrane. We have proposed that membrane insertion is the initial step in this export process. We investigated the interaction of the major inducible hsp from mammalian (Hsp70) and bacterial (DnaK) species with liposomes. We found that mammalian Hsp70 displayed a high specificity for negatively charged phospholipids, such as phosphatidyl serine, whereas DnaK interacted with all lipids tested regardless of the charge. Both proteins were inserted into the lipid bilayer as demonstrated by resistance to acid or basic washes that was confirmed by partial protection from proteolytic cleavage. Several regions of mammalian Hsp70 were inserted into the membrane with a small portion of the N-terminus end exposed to the outer phase of the liposome. In contrast, the N-terminus end of DnaK was inserted into the membrane, exposing the C-terminus end outside the liposome. Mammalian Hsp70 was found to make high oligomeric complexes upon insertion into the membranes whereas DnaK only formed dimers within the lipid bilayer. These observations suggest that both Hsp70s interact with lipids, but mammalian Hsp70 displays a high degree of specificity and structure as compared with the bacterial form.
Collapse
Affiliation(s)
- Victor Lopez
- Initiative for Maximizing Student Development (IMSD) Program, La Jolla, CA, USA
| | - David M Cauvi
- Department of Surgery, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | - Antonio De Maio
- Department of Surgery, School of Medicine, University of California San Diego, La Jolla, CA, USA.
- Center for Investigations of Health and Education Disparities, La Jolla, CA, USA.
- Department of Neuroscience, School of Medicine, University of California San Diego, La Jolla, CA, USA.
- University of California San Diego, 9500 Gilman Drive, #0739, La Jolla, CA, 92093-0739, USA.
| |
Collapse
|
62
|
Radons J. The human HSP70 family of chaperones: where do we stand? Cell Stress Chaperones 2016; 21:379-404. [PMID: 26865365 PMCID: PMC4837186 DOI: 10.1007/s12192-016-0676-6] [Citation(s) in RCA: 377] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/29/2016] [Accepted: 01/29/2016] [Indexed: 01/23/2023] Open
Abstract
The 70-kDa heat shock protein (HSP70) family of molecular chaperones represents one of the most ubiquitous classes of chaperones and is highly conserved in all organisms. Members of the HSP70 family control all aspects of cellular proteostasis such as nascent protein chain folding, protein import into organelles, recovering of proteins from aggregation, and assembly of multi-protein complexes. These chaperones augment organismal survival and longevity in the face of proteotoxic stress by enhancing cell viability and facilitating protein damage repair. Extracellular HSP70s have a number of cytoprotective and immunomodulatory functions, the latter either in the context of facilitating the cross-presentation of immunogenic peptides via major histocompatibility complex (MHC) antigens or in the context of acting as "chaperokines" or stimulators of innate immune responses. Studies have linked the expression of HSP70s to several types of carcinoma, with Hsp70 expression being associated with therapeutic resistance, metastasis, and poor clinical outcome. In malignantly transformed cells, HSP70s protect cells from the proteotoxic stress associated with abnormally rapid proliferation, suppress cellular senescence, and confer resistance to stress-induced apoptosis including protection against cytostatic drugs and radiation therapy. All of the cellular activities of HSP70s depend on their adenosine-5'-triphosphate (ATP)-regulated ability to interact with exposed hydrophobic surfaces of proteins. ATP hydrolysis and adenosine diphosphate (ADP)/ATP exchange are key events for substrate binding and Hsp70 release during folding of nascent polypeptides. Several proteins that bind to distinct subdomains of Hsp70 and consequently modulate the activity of the chaperone have been identified as HSP70 co-chaperones. This review focuses on the regulation, function, and relevance of the molecular Hsp70 chaperone machinery to disease and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Jürgen Radons
- Scientific Consulting International, Mühldorfer Str. 64, 84503, Altötting, Germany.
| |
Collapse
|
63
|
Bromberg Z, Weiss Y. The Role of the Membrane-Initiated Heat Shock Response in Cancer. Front Mol Biosci 2016; 3:12. [PMID: 27200359 PMCID: PMC4847117 DOI: 10.3389/fmolb.2016.00012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 03/18/2016] [Indexed: 01/23/2023] Open
Abstract
The heat shock response (HSR) is a cellular response to diverse environmental and physiological stressors resulting in the induction of genes encoding molecular chaperones, proteases, and other proteins that are essential for protection and recovery from cellular damage. Since different perturbations cause accumulation of misfolded proteins, cells frequently encounter fluctuations in the environment which alter proteostasis. Since tumor cells use their natural adaptive mechanism of coping with stress and misfolded proteins, in recent years, the proteostasis network became a promising target for anti-tumor therapy. The membrane is the first to be affected by heat shock and therefore may be the first one to sense heat shock. The membrane also connects between the extracellular and the intracellular signals. Hence, there is a “cross talk” between the HSR and the membranes since heat shock can induce changes in the fluidity of membranes, leading to membrane lipid remodeling that occurs in several diseases such as cancer. During the last decade, a new possible therapy has emerged in which an external molecule is used that could induce membrane lipid re-organization. Since at the moment there are very few substances that regulate the HSR effectively, an alternative way has been searched to modulate chaperone activities through the plasma membrane. Recently, we suggested that the use of the membrane Transient Receptor Potential Vanilloid-1 (TRPV1) modulators regulated the HSR in cancer cells. However, the primary targets of the signal transduction pathway are yet un-known. This review provides an overview of the current literature regarding the role of HSR in membrane remodeling in cancer since a deep understanding of the membrane biology in cancer and the membrane heat sensing pathway is essential to design novel efficient therapies.
Collapse
Affiliation(s)
- Zohar Bromberg
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University School of Medicine Jerusalem, Israel
| | - Yoram Weiss
- Hadassah Medical Organization Jerusalem, Israel
| |
Collapse
|
64
|
Multhoff G, Habl G, Combs SE. Rationale of hyperthermia for radio(chemo)therapy and immune responses in patients with bladder cancer: Biological concepts, clinical data, interdisciplinary treatment decisions and biological tumour imaging. Int J Hyperthermia 2016; 32:455-63. [PMID: 27050781 DOI: 10.3109/02656736.2016.1152632] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Bladder cancer, the most common tumour of the urinary tract, ranks fifth among all tumour entities. While local treatment or intravesical instillation of bacillus Calmette-Guerin (BCG) provides a treatment option for non-muscle invasive bladder cancer of low grade, surgery or radio(chemo)therapy (RT) are frequently applied in high grade tumours. It remains a matter of debate whether surgery or RT is superior with respect to clinical outcome and quality of life. Surgical resection of bladder cancer can be limited by acute side effects, whereas, RT, which offers a non-invasive treatment option with organ- and functional conservation, can cause long-term side effects. Bladder toxicity by RT mainly depends on the total irradiation dose, fraction size and tumour volume. Therefore, novel approaches are needed to improve clinical outcome. Local tumour hyperthermia is currently used either as an ablation therapy or in combination with RT to enhance anti-tumour effects. In combination with RT an increase of the temperature in the bladder stimulates the local blood flow and as a result can improve the oxygenation state of the tumour, which in turn enhances radiation-induced DNA damage and drug toxicity. Hyperthermia at high temperatures can also directly kill cells, particularly in tumour areas which are poorly perfused, hypoxic or have a low tissue pH. This review summarises current knowledge relating to the role of hyperthermia in RT to treat bladder cancer, the induction and manifestation of immunological responses induced by hyperthermia, and the utilisation of the stress proteins as tumour-specific targets for tumour detection and monitoring of therapeutic outcome.
Collapse
Affiliation(s)
- Gabriele Multhoff
- a Department of Radiation Oncology , Technische Universität München, Klinikum rechts der Isar , Munich ;,b Department of Innovative Radiation Oncology, Department of Radiation Sciences , Helmholtz Zentrum München , Neuherberg , Germany
| | - Gregor Habl
- a Department of Radiation Oncology , Technische Universität München, Klinikum rechts der Isar , Munich
| | - Stephanie E Combs
- a Department of Radiation Oncology , Technische Universität München, Klinikum rechts der Isar , Munich ;,b Department of Innovative Radiation Oncology, Department of Radiation Sciences , Helmholtz Zentrum München , Neuherberg , Germany
| |
Collapse
|
65
|
McCallister C, Kdeiss B, Oliverio R, Nikolaidis N. Characterization of the binding between a 70-kDa heat shock protein, HspA1A, and phosphoinositides. Biochem Biophys Res Commun 2016; 472:270-5. [PMID: 26923070 DOI: 10.1016/j.bbrc.2016.02.103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 02/23/2016] [Indexed: 02/04/2023]
Abstract
HspA1A, a seventy-kilodalton heat shock protein, binds to specific anionic lipids and this interaction regulates important physiological phenomena like apoptosis, tumor growth, and lysosomal rescue. However, whether HspA1A binds to phosphoinositides has yet to be established and quantified. Therefore, in this study, we determined the binding affinity of HspA1A to several phosphoinositides and characterized five aspects of their molecular interaction. First, we established that HspA1A binds phosphatidylinositol monophosphates with higher affinity than di- and triphosphorylated inositides. Second, using high concentrations of potassium we found that HSPA1A embeds within the lipid bilayer of all phosphoinositides tested. However, the effects of the high salt concentrations were significantly different between the different phosphoinositides. Third, using calcium and reaction buffers equilibrated at different pH values we found that these differentially affected HspA1A-phosphoinositide binding, revealing a lipid-specific pattern of binding. Fourth, by assessing the binding properties of the two HspA1A domains, the nucleotide-binding domain and the substrate-binding domain, we determined that in most cases the full-length protein is necessary for binding to phosphoinositides. Fifth, by including in the reactions nucleotides and protein substrates we determined that they minimally and differentially affected phosphoinositide-binding. Collectively, these findings strongly suggest that the HspA1A-phosphoinositide binding is complex yet specific, is mediated by both electrostatic and hydrophobic interactions, is not related to the lipid-head charge, and depends on the physicochemical properties of the lipid.
Collapse
Affiliation(s)
- Chelsea McCallister
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, USA
| | - Brianna Kdeiss
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, USA
| | - Ryan Oliverio
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, USA
| | - Nikolas Nikolaidis
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, USA.
| |
Collapse
|
66
|
Müller G. Personalized Diagnosis and Therapy. DRUG DISCOVERY AND EVALUATION: PHARMACOLOGICAL ASSAYS 2016:3167-3284. [DOI: 10.1007/978-3-319-05392-9_152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
67
|
McCallister C, Kdeiss B, Nikolaidis N. Biochemical characterization of the interaction between HspA1A and phospholipids. Cell Stress Chaperones 2016; 21:41-53. [PMID: 26342809 PMCID: PMC4679732 DOI: 10.1007/s12192-015-0636-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 08/25/2015] [Accepted: 08/31/2015] [Indexed: 01/15/2023] Open
Abstract
Seventy-kilodalton heat shock proteins (Hsp70s) are molecular chaperones essential for maintaining cellular homeostasis. Apart from their indispensable roles in protein homeostasis, specific Hsp70s localize at the plasma membrane and bind to specific lipids. The interaction of Hsp70s with lipids has direct physiological outcomes including lysosomal rescue, microautophagy, and promotion of cell apoptosis. Despite these essential functions, the Hsp70-lipid interactions remain largely uncharacterized. In this study, we characterized the interaction of HspA1A, an inducible Hsp70, with five phospholipids. We first used high concentrations of potassium and established that HspA1A embeds in membranes when bound to all anionic lipids tested. Furthermore, we found that protein insertion is enhanced by increasing the saturation level of the lipids. Next, we determined that the nucleotide-binding domain (NBD) of the protein binds to lipids quantitatively more than the substrate-binding domain (SBD). However, for all lipids tested, the full-length protein is necessary for embedding. We also used calcium and reaction buffers equilibrated at different pH values and determined that electrostatic interactions alone may not fully explain the association of HspA1A with lipids. We then determined that lipid binding is inhibited by nucleotide-binding, but it is unaffected by protein-substrate binding. These results suggest that the HspA1A lipid-association is specific, depends on the physicochemical properties of the lipid, and is mediated by multiple molecular forces. These mechanistic details of the Hsp70-lipid interactions establish a framework of possible physiological functions as they relate to chaperone regulation and localization.
Collapse
Affiliation(s)
- Chelsea McCallister
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA
| | - Brianna Kdeiss
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA
| | - Nikolas Nikolaidis
- Department of Biological Science, Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, California State University, Fullerton, Fullerton, CA, 92834, USA.
| |
Collapse
|
68
|
Shevtsov MA, Nikolaev BP, Ryzhov VA, Yakovleva LY, Marchenko YY, Parr MA, Rolich VI, Mikhrina AL, Dobrodumov AV, Pitkin E, Multhoff G. Ionizing radiation improves glioma-specific targeting of superparamagnetic iron oxide nanoparticles conjugated with cmHsp70.1 monoclonal antibodies (SPION-cmHsp70.1). NANOSCALE 2015; 7:20652-20664. [PMID: 26599206 DOI: 10.1039/c5nr06521f] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The stress-inducible 72 kDa heat shock protein Hsp70 is known to be expressed on the membrane of highly aggressive tumor cells including high-grade gliomas, but not on the corresponding normal cells. Membrane Hsp70 (mHsp70) is rapidly internalized into tumor cells and thus targeting of mHsp70 might provide a promising strategy for theranostics. Superparamagnetic iron oxide nanoparticles (SPIONs) are contrast negative agents that are used for the detection of tumors with MRI. Herein, we conjugated the Hsp70-specific antibody (cmHsp70.1) which is known to recognize mHsp70 to superparamagnetic iron nanoparticles to assess tumor-specific targeting before and after ionizing irradiation. In vitro experiments demonstrated the selectivity of SPION-cmHsp70.1 conjugates to free and mHsp70 in different tumor cell types (C6 glioblastoma, K562 leukemia, HeLa cervix carcinoma) in a dose-dependent manner. High-resolution MRI (11 T) on T(2)-weighted images showed the retention of the conjugates in the C6 glioma model. Accumulation of SPION-cmHsp70.1 nanoparticles in the glioma resulted in a nearly 2-fold drop of T*(2) values in comparison to non-conjugated SPIONs. Biodistribution analysis using NLR-M(2) measurements showed a 7-fold increase in the tumor-to-background (normal brain) uptake ratio of SPION-cmHsp70.1 conjugates in glioma-bearing rats in comparison to SPIONs. This accumulation within Hsp70-positive glioma was further enhanced after a single dose (10 Gy) of ionizing radiation. Elevated accumulation of the magnetic conjugates in the tumor due to radiosensitization proves the combination of radiotherapy and application of Hsp70-targeted agents in brain tumors.
Collapse
Affiliation(s)
- Maxim A Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky ave., 4, St. Petersburg, 194064, Russia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Wang Y, Wang X, Ferrone CR, Schwab JH, Ferrone S. Intracellular antigens as targets for antibody based immunotherapy of malignant diseases. Mol Oncol 2015; 9:1982-93. [PMID: 26597109 DOI: 10.1016/j.molonc.2015.10.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/13/2022] Open
Abstract
This review discusses the potential use of intracellular tumor antigens as targets of antibody-based immunotherapy for the treatment of solid tumors. In addition, it describes the characteristics of the intracellular tumor antigens targeted with antibodies which have been described in the literature and have been identified in the authors' laboratory. Finally, the mechanism underlying the trafficking of the intracellular tumor antigens to the plasma membrane of tumor cells are reviewed.
Collapse
Affiliation(s)
- Yangyang Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Xinhui Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Joseph H Schwab
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States; Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States.
| |
Collapse
|
70
|
Cell surface localised Hsp70 is a cancer specific regulator of clathrin-independent endocytosis. FEBS Lett 2015; 589:2747-53. [PMID: 26257049 DOI: 10.1016/j.febslet.2015.07.037] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 07/01/2015] [Accepted: 07/19/2015] [Indexed: 11/23/2022]
Abstract
The stress inducible heat shock protein 70 (Hsp70) is present specifically on the tumour cell surface yet without a pro-tumour function revealed. We show here that cell surface localised Hsp70 (sHsp70) supports clathrin-independent endocytosis (CIE) in melanoma models. Remarkably, ability of Hsp70 to cluster on lipid rafts in vitro correlated with larger nano-domain sizes of sHsp70 in high sHsp70 expressing cell membranes. Interfering with Hsp70 oligomerisation impaired sHsp70-mediated facilitation of endocytosis. Altogether our findings suggest that a sub-fraction of sHsp70 co-localising with lipid rafts enhances CIE through oligomerisation and clustering. Targeting or utilising this tumour specific mechanism may represent an additional benefit for anti-cancer therapy.
Collapse
|
71
|
Murakami N, Kühnel A, Schmid TE, Ilicic K, Stangl S, Braun IS, Gehrmann M, Molls M, Itami J, Multhoff G. Role of membrane Hsp70 in radiation sensitivity of tumor cells. Radiat Oncol 2015. [PMID: 26197988 PMCID: PMC4511458 DOI: 10.1186/s13014-015-0461-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The major stress-inducible heat shock protein 70 (Hsp70) is frequently overexpressed in the cytosol and integrated in the plasma membrane of tumor cells via lipid anchorage. Following stress such as non-lethal irradiation Hsp70 synthesis is up-regulated. Intracellular located Hsp70 is known to exert cytoprotective properties, however, less is known about membrane (m)Hsp70. Herein, we investigate the role of mHsp70 in the sensitivity towards irradiation in tumor sublines that differ in their cytosolic and/or mHsp70 levels. METHODS The isogenic human colon carcinoma sublines CX(+) with stable high and CX(-) with stable low expression of mHsp70 were generated by fluorescence activated cell sorting, the mouse mammary carcinoma sublines 4 T1 (4 T1 ctrl) and Hsp70 knock-down (4 T1 Hsp70 KD) were produced using the CRISPR/Cas9 system, and the Hsp70 down-regulation in human lung carcinoma sublines H1339 ctrl/H1339 HSF-1 KD and EPLC-272H ctrl/EPLC-272H HSF-1 KD was achieved by small interfering (si)RNA against Heat shock factor 1 (HSF-1). Cytosolic and mHsp70 was quantified by Western blot analysis/ELISA and flow cytometry; double strand breaks (DSBs) and apoptosis were measured by flow cytometry using antibodies against γH2AX and real-time PCR (RT-PCR) using primers and antibodies directed against apoptosis related genes; and radiation sensitivity was determined using clonogenic cell surviving assays. RESULTS CX(+)/CX(-) tumor cells exhibited similar cytosolic but differed significantly in their mHsp70 levels, 4 T1 ctrl/4 T1 Hsp70 KD cells showed significant differences in their cytosolic and mHsp70 levels and H1339 ctrl/H1339 HSF-1 KD and EPLC-272H ctrl/EPLC-272H HSF-1 KD lung carcinoma cell sublines had similar mHsp70 but significantly different cytosolic Hsp70 levels. γH2AX was significantly up-regulated in irradiated CX(-) and 4 T1 Hsp70 KD with low basal mHsp70 levels, but not in their mHsp70 high expressing counterparts, irrespectively of their cytosolic Hsp70 content. After irradiation γH2AX, Caspase 3/7 and Annexin V were up-regulated in the lung carcinoma sublines, but no significant differences were observed in H1339 ctrl/H1339 HSF-1 KD, and EPLC-272H ctrl/EPLC-272H HSF-1 KD that exhibit identical mHsp70 but different cytosolic Hsp70 levels. Clonogenic cell survival was significantly lower in CX(-) and 4 T1 Hsp70 KD cells with low mHsp70 expression, than in CX+ and 4 T1 ctrl cells, whereas no difference in clonogenic cell survival was observed in H1339 ctrl/H1339 HSF-1 KD and EPLC-272H ctrl/ EPLC-272H HSF-1 KD sublines with identical mHsp70 but different cytosolic Hsp70 levels. CONCLUSION In summary, our results indicate that mHsp70 has an impact on radiation resistance.
Collapse
Affiliation(s)
- Naoya Murakami
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Department of Radiation Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Annett Kühnel
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Thomas E Schmid
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Katarina Ilicic
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Stefan Stangl
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Isabella S Braun
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Mathias Gehrmann
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Michael Molls
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Jun Itami
- Department of Radiation Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany. .,Clinical Cooperation Group - Innate Immunity in Tumor Biology, Institute of Biomedical Imaging (IBMI), Helmholtz Zentrum München, Munich, Germany.
| |
Collapse
|
72
|
Sulfatide-Hsp70 interaction promotes Hsp70 clustering and stabilizes binding to unfolded protein. Biomolecules 2015; 5:958-73. [PMID: 25989600 PMCID: PMC4496704 DOI: 10.3390/biom5020958] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 04/24/2015] [Accepted: 05/07/2015] [Indexed: 01/01/2023] Open
Abstract
The 70-kDa heat shock protein (Hsp70), one of the major stress-inducible molecular chaperones, is localized not only in the cytosol, but also in extracellular milieu in mammals. Hsp70 interacts with various cell surface glycolipids including sulfatide (3'-sulfogalactosphingolipid). However, the molecular mechanism, as well as the biological relevance, underlying the glycolipid-Hsp70 interaction is unknown. Here we report that sulfatide promotes Hsp70 oligomerization through the N-terminal ATPase domain, which stabilizes the binding of Hsp70 to unfolded protein in vitro. We find that the Hsp70 oligomer has apparent molecular masses ranging from 440 kDa to greater than 669 kDa. The C-terminal peptide-binding domain is dispensable for the sulfatide-induced oligomer formation. The oligomer formation is impaired in the presence of ATP, while the Hsp70 oligomer, once formed, is unable to bind to ATP. These results suggest that sulfatide locks Hsp70 in a high-affinity state to unfolded proteins by clustering the peptide-binding domain and blocking the binding to ATP that induces the dissociation of Hsp70 from protein substrates.
Collapse
|
73
|
Specht HM, Ahrens N, Blankenstein C, Duell T, Fietkau R, Gaipl US, Günther C, Gunther S, Habl G, Hautmann H, Hautmann M, Huber RM, Molls M, Offner R, Rödel C, Rödel F, Schütz M, Combs SE, Multhoff G. Heat Shock Protein 70 (Hsp70) Peptide Activated Natural Killer (NK) Cells for the Treatment of Patients with Non-Small Cell Lung Cancer (NSCLC) after Radiochemotherapy (RCTx) - From Preclinical Studies to a Clinical Phase II Trial. Front Immunol 2015; 6:162. [PMID: 25926832 PMCID: PMC4397864 DOI: 10.3389/fimmu.2015.00162] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 03/25/2015] [Indexed: 11/23/2022] Open
Abstract
Heat shock protein 70 (Hsp70) is frequently overexpressed in tumor cells. An unusual cell surface localization could be demonstrated on a large variety of solid tumors including lung, colorectal, breast, squamous cell carcinomas of the head and neck, prostate and pancreatic carcinomas, glioblastomas, sarcomas and hematological malignancies, but not on corresponding normal tissues. A membrane (m)Hsp70-positive phenotype can be determined either directly on single cell suspensions of tumor biopsies by flow cytometry using cmHsp70.1 monoclonal antibody or indirectly in the serum of patients using a novel lipHsp70 ELISA. A mHsp70-positive tumor phenotype has been associated with highly aggressive tumors, causing invasion and metastases and resistance to cell death. However, natural killer (NK), but not T cells were found to kill mHsp70-positive tumor cells after activation with a naturally occurring Hsp70 peptide (TKD) plus low dose IL-2 (TKD/IL-2). Safety and tolerability of ex vivo TKD/IL-2 stimulated, autologous NK cells has been demonstrated in patients with metastasized colorectal and non-small cell lung cancer (NSCLC) in a phase I clinical trial. Based on promising clinical results of the previous study, a phase II randomized clinical study was initiated in 2014. The primary objective of this multicenter proof-of-concept trial is to examine whether an adjuvant treatment of NSCLC patients after platinum-based radiochemotherapy (RCTx) with TKD/IL-2 activated, autologous NK cells is clinically effective. As a mHsp70-positive tumor phenotype is associated with poor clinical outcome only mHsp70-positive tumor patients will be recruited into the trial. The primary endpoint of this study will be the comparison of the progression-free survival of patients treated with ex vivo activated NK cells compared to patients who were treated with RCTx alone. As secondary endpoints overall survival, toxicity, quality-of-life, and biological responses will be determined in both study groups.
Collapse
Affiliation(s)
- Hanno M Specht
- Radiation Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| | - Norbert Ahrens
- Transfusion Medicine, Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg , Regensburg , Germany
| | - Christiane Blankenstein
- Münchner Studienzentrum (MSZ), Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| | - Thomas Duell
- Thoracic Oncology, Asklepios Lungenfachkliniken , Munich , Germany
| | - Rainer Fietkau
- Radiation Oncology, University Hospital Erlangen , Erlangen , Germany
| | - Udo S Gaipl
- Radiation Oncology, University Hospital Erlangen , Erlangen , Germany
| | | | - Sophie Gunther
- Radiation Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| | - Gregor Habl
- Radiation Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| | - Hubert Hautmann
- Thoracic Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| | - Matthias Hautmann
- Radiation Oncology, University Hospital Regensburg , Regensburg , Germany
| | - Rudolf Maria Huber
- Thoracic Oncology, Department of Medicine, University of Munich , Munich , Germany
| | - Michael Molls
- Radiation Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| | - Robert Offner
- Transfusion Medicine, Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg , Regensburg , Germany
| | - Claus Rödel
- Radiation Oncology, University Hospital Frankfurt , Frankfurt , Germany
| | - Franz Rödel
- Radiation Oncology, University Hospital Frankfurt , Frankfurt , Germany
| | - Martin Schütz
- Thoracic Oncology, Klinikum Bogenhausen , Munich , Germany
| | - Stephanie E Combs
- Radiation Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| | - Gabriele Multhoff
- Radiation Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany ; Institute of Biological Molecular Imaging, Helmholtz Zentrum München , Munich , Germany
| |
Collapse
|
74
|
Functional diversification and specialization of cytosolic 70-kDa heat shock proteins. Sci Rep 2015; 5:9363. [PMID: 25791537 PMCID: PMC4366816 DOI: 10.1038/srep09363] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/02/2015] [Indexed: 02/04/2023] Open
Abstract
A fundamental question in molecular evolution is how protein functional differentiation alters the ability of cells and organisms to cope with stress and survive. To answer this question we used two paralogous Hsp70s from mouse and explored whether these highly similar cytosolic molecular chaperones, which apart their temporal expression have been considered functionally interchangeable, are differentiated with respect to their lipid-binding function. We demonstrate that the two proteins bind to diverse lipids with different affinities and therefore are functionally specialized. The observed lipid-binding patterns may be related with the ability of both Hsp70s to induce cell death by binding to a particular plasma-membrane lipid, and the potential of only one of them to promote cell survival by binding to a specific lysosomal-membrane lipid. These observations reveal that two seemingly identical proteins differentially modulate cellular adaptation and survival by having acquired specialized functions via sequence divergence. Therefore, this study provides an evolutionary paradigm, where promiscuity, specificity, sub- and neo-functionalization orchestrate one of the most conserved systems in nature, the cellular stress-response.
Collapse
|
75
|
Gaipl US, Multhoff G, Scheithauer H, Lauber K, Hehlgans S, Frey B, Rödel F. Kill and spread the word: stimulation of antitumor immune responses in the context of radiotherapy. Immunotherapy 2015; 6:597-610. [PMID: 24896628 DOI: 10.2217/imt.14.38] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Besides the direct, targeted effects of ionizing irradiation (x-ray) on cancer cells, namely DNA damage and cell death induction, indirect, nontargeted ones exist, which are mediated in large part by the immune system. Immunogenic forms of tumor cell death induced by x-ray, including immune modulating danger signals like the heat shock protein 70, adenosine triphosphate, and high-mobility group box 1 protein are presented. Further, antitumor effects exerted by cells of the innate (natural killer cells) as well as adaptive immune system (T cells activated by dendritic cells) are outlined. Tumor cell death inhibiting molecules such as survivin are introduced as suitable target for molecularly tailored therapies in combination with x-ray. Finally, reasonable combinations of immune therapies with radiotherapy are discussed.
Collapse
Affiliation(s)
- Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | | | | | | | | | | | | |
Collapse
|
76
|
Multhoff G, Pockley AG, Schmid TE, Schilling D. The role of heat shock protein 70 (Hsp70) in radiation-induced immunomodulation. Cancer Lett 2015; 368:179-84. [PMID: 25681671 DOI: 10.1016/j.canlet.2015.02.013] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/04/2015] [Accepted: 02/07/2015] [Indexed: 02/08/2023]
Abstract
Despite enormous progress in radiation technologies (high precision image-guided irradiation, proton irradiation, heavy ion irradiation) and radiotherapeutic concepts (hypofractionated irradiation schemes), the clinical outcome of radiotherapy in locally advanced and metastasized tumors and in hypoxic tumors which are radiation-resistant remains unsatisfactory. Given their key influence on a number of biological and immunological parameters, this article considers the influence of irradiation-induced stress proteins on radiation-induced immunomodulation. Depending on its location, the major stress-inducible Heat shock protein 70 (Hsp70) has been found to fulfill multiple roles. On the one hand, increased intracellular Hsp70 levels have been found to play a key role in the recovery from stress such as radio(chemo)therapy, and on the other hand extracellular Hsp70 proteins are potent stimulators of the innate immune system and mediators of anti-tumor immunity. Furthermore, if loaded with tumor-derived peptides, members of the Heat Shock Protein 70 (HSP70) and 90 (HSP90) families can stimulate the adaptive immune system via antigen cross-presentation. An irradiation-induced enhancement of the selective expression of a membrane form of Hsp70 on the surface of tumor cells which can act as a recognition structure for activated NK cells might have significant clinical relevance, in that the outcome of irradiation therapy for advanced tumors could be improved by combining it with cell-based and other immunotherapies that target this membrane form of Hsp70.
Collapse
Affiliation(s)
- Gabriele Multhoff
- Department of Radiation Oncology, Technische Universität München, Klinikum rechts der Isar, Munich, Germany; Helmholtz Center Munich, German Research Center for Environmental Health, CCG - "Innate Immunity in Tumor Biology", Munich, Germany.
| | - Alan G Pockley
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Thomas E Schmid
- Department of Radiation Oncology, Technische Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Daniela Schilling
- Department of Radiation Oncology, Technische Universität München, Klinikum rechts der Isar, Munich, Germany
| |
Collapse
|
77
|
Rödel F, Frey B, Multhoff G, Gaipl U. Contribution of the immune system to bystander and non-targeted effects of ionizing radiation. Cancer Lett 2015; 356:105-13. [DOI: 10.1016/j.canlet.2013.09.015] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 08/13/2013] [Accepted: 09/11/2013] [Indexed: 12/21/2022]
|
78
|
Gehrmann M, Stangl S, Foulds GA, Oellinger R, Breuninger S, Rad R, Pockley AG, Multhoff G. Tumor imaging and targeting potential of an Hsp70-derived 14-mer peptide. PLoS One 2014; 9:e105344. [PMID: 25165986 PMCID: PMC4148261 DOI: 10.1371/journal.pone.0105344] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/21/2014] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND We have previously used a unique mouse monoclonal antibody cmHsp70.1 to demonstrate the selective presence of a membrane-bound form of Hsp70 (memHsp70) on a variety of leukemia cells and on single cell suspensions derived from solid tumors of different entities, but not on non-transformed cells or cells from corresponding 'healthy' tissue. This antibody can be used to image tumors in vivo and target them for antibody-dependent cellular cytotoxicity. Tumor-specific expression of memHsp70 therefore has the potential to be exploited for theranostic purposes. Given the advantages of peptides as imaging and targeting agents, this study assessed whether a 14-mer tumor penetrating peptide (TPP; TKDNNLLGRFELSG), the sequence of which is derived from the oligomerization domain of Hsp70 which is expressed on the cell surface of tumor cells, can also be used for targeting membrane Hsp70 positive (memHsp70+) tumor cells, in vitro. METHODOLOGY/PRINCIPAL FINDINGS The specificity of carboxy-fluorescein (CF-) labeled TPP (TPP) to Hsp70 was proven in an Hsp70 knockout mammary tumor cell system. TPP specifically binds to different memHsp70+ mouse and human tumor cell lines and is rapidly taken up via endosomes. Two to four-fold higher levels of CF-labeled TPP were detected in MCF7 (82% memHsp70+) and MDA-MB-231 (75% memHsp70+) cells compared to T47D cells (29% memHsp70+) that exhibit a lower Hsp70 membrane positivity. After 90 min incubation, TPP co-localized with mitochondrial membranes in memHsp70+ tumors. Although there was no evidence that any given vesicle population was specifically localized, fluorophore-labeled cmHsp70.1 antibody and TPP preferentially accumulated in the proximity of the adherent surface of cultured cells. These findings suggest a potential association between membrane Hsp70 expression and cytoskeletal elements that are involved in adherence, the establishment of intercellular synapses and/or membrane reorganization. CONCLUSIONS/SIGNIFICANCE This study demonstrates the specific binding and rapid internalization of TPP by tumor cells with a memHsp70+ phenotype. TPP might therefore have potential for targeting and imaging the large proportion of tumors (∼50%) that express memHsp70.
Collapse
Affiliation(s)
- Mathias Gehrmann
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Stefan Stangl
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Gemma A. Foulds
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, United Kingdom
| | - Rupert Oellinger
- Medical Department II, Translational Gastroenterological Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Stephanie Breuninger
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Roland Rad
- Medical Department II, Translational Gastroenterological Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Alan G. Pockley
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, United Kingdom
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Clinical Cooperation Group (CCG) ‘‘Innate Immunity in Tumor Biology’’, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Munich, Germany
| |
Collapse
|
79
|
Mahalka AK, Kirkegaard T, Jukola LT, Jäättelä M, Kinnunen PK. Human heat shock protein 70 (Hsp70) as a peripheral membrane protein. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:1344-61. [DOI: 10.1016/j.bbamem.2014.01.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 01/13/2014] [Accepted: 01/17/2014] [Indexed: 11/28/2022]
|
80
|
Shevtsov MA, Yakovleva LY, Nikolaev BP, Marchenko YY, Dobrodumov AV, Onokhin KV, Onokhina YS, Selkov SA, Mikhrina AL, Guzhova IV, Martynova MG, Bystrova OA, Ischenko AM, Margulis BA. Tumor targeting using magnetic nanoparticle Hsp70 conjugate in a model of C6 glioma. Neuro Oncol 2013; 16:38-49. [PMID: 24305705 DOI: 10.1093/neuonc/not141] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Superparamagnetic iron oxide nanoparticles (SPIONs), due to their unique magnetic properties, have the ability to function both as magnetic resonance (MR) contrast agents, and can be used for thermotherapy. SPIONs conjugated to the heat shock protein Hsp70 that selectively binds to the CD40 receptor present on glioma cells, could be used for MR contrast enhancement of experimental C6 glioma. METHODS The magnetic properties of the Hsp70-SPIONs were measured by NMR relaxometry method. The uptake of nanoparticles was assessed on the C6 glioma cells by confocal and electron microscopes. The tumor selectivity of Hsp70-SPIONs being intravenously administered was analyzed in the experimental model of C6 glioma in the MRI scanner. RESULTS Hsp70-SPIONs relaxivity corresponded to the properties of negative contrast agents with a hypointensive change of resonance signal in MR imaging. A significant accumulation of the Hsp70-SPIONs but not the non-conjugated nanoparticles was observed by confocal microscopy within C6 cells. Negative contrast tumor enhancement in the T2-weighted MR images was higher in the case of Hsp70-SPIONs in comparison to non-modified SPIONs. Histological analysis of the brain sections confirmed the retention of the Hsp70-SPIONs in the glioma tumor but not in the adjacent normal brain tissues. CONCLUSION The study demonstrated that Hsp70-SPION conjugate intravenously administered in C6 glioma model accumulated in the tumors and enhanced the contrast of their MR images.
Collapse
Affiliation(s)
- Maxim A Shevtsov
- Corresponding author: Maxim A. Shevtsov, MD, PhD, Laboratory of Cell Protection Mechanisms, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Hajj GNM, Arantes CP, Dias MVS, Roffé M, Costa-Silva B, Lopes MH, Porto-Carreiro I, Rabachini T, Lima FR, Beraldo FH, Prado MMA, Linden R, Martins VR. The unconventional secretion of stress-inducible protein 1 by a heterogeneous population of extracellular vesicles. Cell Mol Life Sci 2013; 70:3211-27. [PMID: 23543276 PMCID: PMC11113396 DOI: 10.1007/s00018-013-1328-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 03/15/2013] [Accepted: 03/18/2013] [Indexed: 01/03/2023]
Abstract
The co-chaperone stress-inducible protein 1 (STI1) is released by astrocytes, and has important neurotrophic properties upon binding to prion protein (PrP(C)). However, STI1 lacks a signal peptide and pharmacological approaches pointed that it does not follow a classical secretion mechanism. Ultracentrifugation, size exclusion chromatography, electron microscopy, vesicle labeling, and particle tracking analysis were used to identify three major types of extracellular vesicles (EVs) released from astrocytes with sizes ranging from 20-50, 100-200, and 300-400 nm. These EVs carry STI1 and present many exosomal markers, even though only a subpopulation had the typical exosomal morphology. The only protein, from those evaluated here, present exclusively in vesicles that have exosomal morphology was PrP(C). STI1 partially co-localized with Rab5 and Rab7 in endosomal compartments, and a dominant-negative for vacuolar protein sorting 4A (VPS4A), required for formation of multivesicular bodies (MVBs), impaired EV and STI1 release. Flow cytometry and PK digestion demonstrated that STI1 localized to the outer leaflet of EVs, and its association with EVs greatly increased STI1 activity upon PrP(C)-dependent neuronal signaling. These results indicate that astrocytes secrete a diverse population of EVs derived from MVBs that contain STI1 and suggest that the interaction between EVs and neuronal surface components enhances STI1-PrP(C) signaling.
Collapse
Affiliation(s)
- Glaucia N. M. Hajj
- International Research Center, A.C. Camargo Hospital, Rua Taguá 540, São Paulo, 01508-010 Brazil
- National Institute for Translational Neuroscience and National Institute of Oncogenomics, São Paulo, Brazil
| | - Camila P. Arantes
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, Brazil
| | - Marcos Vinicios Salles Dias
- International Research Center, A.C. Camargo Hospital, Rua Taguá 540, São Paulo, 01508-010 Brazil
- National Institute for Translational Neuroscience and National Institute of Oncogenomics, São Paulo, Brazil
| | - Martín Roffé
- International Research Center, A.C. Camargo Hospital, Rua Taguá 540, São Paulo, 01508-010 Brazil
- National Institute for Translational Neuroscience and National Institute of Oncogenomics, São Paulo, Brazil
| | - Bruno Costa-Silva
- International Research Center, A.C. Camargo Hospital, Rua Taguá 540, São Paulo, 01508-010 Brazil
- National Institute for Translational Neuroscience and National Institute of Oncogenomics, São Paulo, Brazil
| | - Marilene H. Lopes
- Department of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Isabel Porto-Carreiro
- Instituto de Biofisica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Flávia R. Lima
- Instituto de Ciências Biomédicas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flávio H. Beraldo
- Department of Anatomy and Cell Biology and Department of Physiology and Pharmacology, Robarts Research Institute, University of Western Ontario, London, Canada
| | - Marco M. A. Prado
- Department of Anatomy and Cell Biology and Department of Physiology and Pharmacology, Robarts Research Institute, University of Western Ontario, London, Canada
| | - Rafael Linden
- Instituto de Biofisica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vilma R. Martins
- International Research Center, A.C. Camargo Hospital, Rua Taguá 540, São Paulo, 01508-010 Brazil
- National Institute for Translational Neuroscience and National Institute of Oncogenomics, São Paulo, Brazil
| |
Collapse
|
82
|
Leng X, Wang X, Pang W, Zhan R, Zhang Z, Wang L, Gao X, Qian L. Evidence of a role for both anti-Hsp70 antibody and endothelial surface membrane Hsp70 in atherosclerosis. Cell Stress Chaperones 2013; 18:483-93. [PMID: 23334859 PMCID: PMC3682019 DOI: 10.1007/s12192-013-0404-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2011] [Revised: 12/27/2012] [Accepted: 01/04/2013] [Indexed: 10/27/2022] Open
Abstract
Although previous studies have shown that autoantigens such as Hsps have been implicated by induction of an autoimmune process in the development of atherosclerosis, the exact role of anti-Hsp70 antibody in atherosclerosis is unknown. In the present study, the levels of anti-Hsp70 autoantibodies and oxidized low density lipoprotein (OxLDL) were all significantly increased, and they were strongly correlated in an atherosclerosis model. After the endothelial cells were incubated with 20 μg/mL OxLDL for 12 h at 37 °C and followed by 90 min recovery, Hsp70 positive staining of OxLDL-treated endothelial cells was observed on the cell surface in immunostaining and flow cytometric analysis. This membrane Hsp70 was not from culture supernatant Hsp70 and binding of extracellular Hsp70 but was defined as endothelial surface membrane Hsp70. Furthermore, only in the OxLDL-treated group, but not in the untreated group, (51)Cr-labeled endothelial cells were lysed by anti-Hsp70 antibody (BD091, Ig(AS)) in the presence of either complement or peripheral blood mononuclear cells. Control antibodies, including Ig(Nor), mAb to Hsp70 (SPA-810), and mAbs to Factor VIII, α-actin, and CD3 showed no cytotoxic effects. In conclusion, anti-Hsp70 antibodies could be reacting with the endothelial surface membrane Hsp70 induced by OxLDL and were able to mediate endothelial cytotoxicity. There is a possibility that a humoral immune reaction to endothelial surface membrane Hsp70 may play an important role in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Xue Leng
- />Institute of Medical Equipment, Tianjin, 300161 People’s Republic of China
| | - Xinxing Wang
- />Institute of Basic Medical Sciences, Beijing, 100850 People’s Republic of China
| | - Wei Pang
- />Institute of Health & Environmental Medicine, Tianjin, 300050 People’s Republic of China
| | - Rui Zhan
- />Institute of Basic Medical Sciences, Beijing, 100850 People’s Republic of China
| | - Zhiqing Zhang
- />Institute of Health & Environmental Medicine, Tianjin, 300050 People’s Republic of China
| | - Liqun Wang
- />Institute of Health & Environmental Medicine, Tianjin, 300050 People’s Republic of China
| | - Xiujie Gao
- />Institute of Health & Environmental Medicine, Tianjin, 300050 People’s Republic of China
| | - Lingjia Qian
- />Institute of Basic Medical Sciences, Beijing, 100850 People’s Republic of China
| |
Collapse
|
83
|
Foulds GA, Radons J, Kreuzer M, Multhoff G, Pockley AG. Influence of tumors on protective anti-tumor immunity and the effects of irradiation. Front Oncol 2013; 3:14. [PMID: 23378947 PMCID: PMC3561630 DOI: 10.3389/fonc.2013.00014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 01/15/2013] [Indexed: 12/20/2022] Open
Abstract
Innate and adaptive immunity plays important roles in the development and progression of cancer and it is becoming apparent that tumors can influence the induction of potentially protective responses in a number of ways. The prevalence of immunoregulatory T cell populations in the circulation and tumors of patients with cancer is increased and the presence of these cells appears to present a major barrier to the induction of tumor immunity. One aspect of tumor-mediated immunoregulation which has received comparatively little attention is that which is directed toward natural killer (NK) cells, although evidence that the phenotype and function of NK cell populations are modified in patients with cancer is accumulating. Although the precise mechanisms underlying these localized and systemic immunoregulatory effects remain unclear, tumor-derived factors appear, in part at least, to be involved. The effects could be manifested by an altered function and/or via an influence on the migratory properties of individual cell subsets. A better insight into endogenous immunoregulatory mechanisms and the capacity of tumors to modify the phenotype and function of innate and adaptive immune cells might assist the development of new immunotherapeutic approaches and improve the management of patients with cancer. This article reviews current knowledge relating to the influence of tumors on protective anti-tumor immunity and considers the potential influence that radiation-induced effects might have on the prevalence, phenotype, and function of innate and adaptive immune cells in patients with cancer.
Collapse
Affiliation(s)
- Gemma A Foulds
- Department of Oncology, The Medical School, The University of Sheffield Sheffield, UK ; Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München Munich, Germany
| | | | | | | | | |
Collapse
|
84
|
Lauber K, Ernst A, Orth M, Herrmann M, Belka C. Dying cell clearance and its impact on the outcome of tumor radiotherapy. Front Oncol 2012; 2:116. [PMID: 22973558 PMCID: PMC3438527 DOI: 10.3389/fonc.2012.00116] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 08/27/2012] [Indexed: 12/29/2022] Open
Abstract
The induction of tumor cell death is one of the major goals of radiotherapy and has been considered to be the central determinant of its therapeutic outcome for a long time. However, accumulating evidence suggests that the success of radiotherapy does not only derive from direct cytotoxic effects on the tumor cells alone, but instead might also depend – at least in part – on innate as well as adaptive immune responses, which can particularly target tumor cells that survive local irradiation. The clearance of dying tumor cells by phagocytic cells of the innate immune system represents a crucial step in this scenario. Dendritic cells and macrophages, which engulf, process and present dying tumor cell material to adaptive immune cells, can trigger, skew, or inhibit adaptive immune responses, respectively. In this review we summarize the current knowledge of different forms of cell death induced by ionizing radiation, the multi-step process of dying cell clearance, and its immunological consequences with special regard toward the potential exploitation of these mechanisms for the improvement of tumor radiotherapy.
Collapse
Affiliation(s)
- Kirsten Lauber
- Department of Radiotherapy and Radiation Oncology, Ludwig Maximilian University of Munich Munich, Germany
| | | | | | | | | |
Collapse
|
85
|
Wang R, Liu X, Küster-Schöck E, Fagotto F. Proteomic analysis of differences in ectoderm and mesoderm membranes by DiGE. J Proteome Res 2012; 11:4575-93. [PMID: 22852788 DOI: 10.1021/pr300379m] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ectoderm and mesoderm can be considered as prototypes for epithelial and mesenchymal cell types. These two embryonic tissues display clear differences in adhesive and motility properties, which are phenomenologically well characterized but remain largely unexplored at the molecular level. Because the key downstream regulations must occur at the plasma membrane and in the underlying actin cortical structures, we have set out to compare the protein content of membrane fractions from Xenopus ectoderm and mesoderm tissues using 2-dimensional difference gel electrophoresis (DiGE). We have thus identified several proteins that are enriched in one or the other tissues, including regulators of the cytoskeleton and of cell signaling. This study represents to our knowledge the first attempt to use proteomics specifically targeted to the membrane-cortex compartment of embryonic tissues. The identified components should help unraveling a variety of tissue-specific functions in the embryo.
Collapse
Affiliation(s)
- Renee Wang
- Department of Biology, McGill University, Montreal, Canada
| | | | | | | |
Collapse
|
86
|
Gehrmann M, Stangl S, Kirschner A, Foulds GA, Sievert W, Doß BT, Walch A, Pockley AG, Multhoff G. Immunotherapeutic targeting of membrane Hsp70-expressing tumors using recombinant human granzyme B. PLoS One 2012; 7:e41341. [PMID: 22829941 PMCID: PMC3400620 DOI: 10.1371/journal.pone.0041341] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 06/20/2012] [Indexed: 01/02/2023] Open
Abstract
Background We have previously reported that human recombinant granzyme B (grB) mediates apoptosis in membrane heat shock protein 70 (Hsp70)-positive tumor cells in a perforin-independent manner. Methodology/Principal Findings Optical imaging of uptake kinetics revealed co-localization of grB with recycling endosomes (Rab9/11) as early as 5 min after internalization, with late endosomes (Rab7) after 30 min, and the lysosomal compartment (LAMP1/2) after 60 to 120 min. Active caspase-3-mediated apoptosis was induced in mouse CT26 monolayer cells and 3D tumor spheroids, but not in normal mouse endothelial cells. Granzyme B selectively reduced the proportion of membrane Hsp70-positive cells in CT26 tumor spheroids. Consecutive i.v. injections of recombinant human grB into mice bearing membrane Hsp70-positive CT26 tumors resulted in significant tumor suppression, and a detailed inspection of normal mouse organs revealed that the administration of anti-tumoral concentrations of grB elicited no clinicopathological changes. Conclusions/Significance These findings support the future clinical evaluation of human grB as a potential adjuvant therapeutic agent, especially for treating immunosuppressed patients that bear membrane Hsp70-positive tumors.
Collapse
Affiliation(s)
- Mathias Gehrmann
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, and Clinical Cooperation Group (CCG) “Innate Immunity in Tumor Biology”, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Munich, Germany
| | - Stefan Stangl
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, and Clinical Cooperation Group (CCG) “Innate Immunity in Tumor Biology”, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Munich, Germany
| | - Andreas Kirschner
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, and Clinical Cooperation Group (CCG) “Innate Immunity in Tumor Biology”, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Munich, Germany
| | - Gemma A. Foulds
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, and Clinical Cooperation Group (CCG) “Innate Immunity in Tumor Biology”, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Munich, Germany
- Department of Oncology, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Wolfgang Sievert
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, and Clinical Cooperation Group (CCG) “Innate Immunity in Tumor Biology”, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Munich, Germany
| | - Brigitte T. Doß
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, and Clinical Cooperation Group (CCG) “Innate Immunity in Tumor Biology”, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Munich, Germany
| | - Axel Walch
- Institute of Pathology, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Munich, Germany
| | - Alan G. Pockley
- Department of Oncology, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, and Clinical Cooperation Group (CCG) “Innate Immunity in Tumor Biology”, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Munich, Germany
- * E-mail:
| |
Collapse
|
87
|
Frey B, Weiss EM, Rubner Y, Wunderlich R, Ott OJ, Sauer R, Fietkau R, Gaipl US. Old and new facts about hyperthermia-induced modulations of the immune system. Int J Hyperthermia 2012; 28:528-42. [PMID: 22690925 DOI: 10.3109/02656736.2012.677933] [Citation(s) in RCA: 178] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Hyperthermia (HT) is a potent sensitiser for radiotherapy (RT) and chemotherapy (CT) and has been proven to modulate directly or indirectly cells of the innate and adaptive immune system. We will focus in this article on how anti-tumour immunity can be induced by HT. In contrast to some in vitro assays, in vivo examinations showed that natural killer cells and phagocytes like granulocytes are directly activated against the tumour by HT. Since heat also activates dendritic cells (DCs), HT should be combined with further death stimuli (RT, CT or immune therapy) to allocate tumour antigen, derived from, for example, necrotic tumour cells, for uptake by DCs. We will outline that induction of immunogenic tumour cells and direct tumour cell killing by HT in combination with other therapies contributes to immune activation against the tumour. Studies will be presented showing that non-beneficial effects of HT on immune cells are mostly timely restricted. A special focus is set on immune activation mediated by extracellular present heat shock proteins (HSPs) carrying tumour antigens and further danger signals released by dying tumour cells. Local HT treatment in addition to further stress stimuli exerts abscopal effects and might be considered as in situ tumour vaccination. An increased natural killer (NK) cell activity, lymphocyte infiltration and HSP-mediated induction of immunogenic tumour cells have been observed in patients. Treatments with the addition of HT therefore can be considered as a personalised cancer treatment approach by specifically activating the immune system against the individual unique tumour.
Collapse
Affiliation(s)
- Benjamin Frey
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
88
|
Multhoff G, Radons J. Radiation, inflammation, and immune responses in cancer. Front Oncol 2012; 2:58. [PMID: 22675673 PMCID: PMC3366472 DOI: 10.3389/fonc.2012.00058] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 05/18/2012] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation has emerged as one of the hallmarks of cancer. Inflammation also plays a pivotal role in modulating radiation responsiveness of tumors. As discussed in this review, ionizing radiation (IR) leads to activation of several transcription factors modulating the expression of numerous mediators in tumor cells and cells of the microenvironment promoting cancer development. Novel therapeutic approaches thus aim to interfere with the activity or expression of these factors, either in single-agent or combinatorial treatment or as supplements of the existing therapeutic concepts. Among them, NF-κB, STAT-3, and HIF-1 play a crucial role in radiation-induced inflammatory responses embedded in a complex inflammatory network. A great variety of classical or novel drugs including nutraceuticals such as plant phytochemicals have the capacity to interfere with the inflammatory network in cancer and are considered as putative radiosensitizers. Thus, targeting the inflammatory signaling pathways induced by IR offers the opportunity to improve the clinical outcome of radiation therapy by enhancing radiosensitivity and decreasing putative metabolic effects. Since inflammation and sex steroids also impact tumorigenesis, a therapeutic approach targeting glucocorticoid receptors and radiation-induced production of tumorigenic factors might be effective in sensitizing certain tumors to IR.
Collapse
Affiliation(s)
- Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München Munich, Germany
| | | |
Collapse
|
89
|
Horváth I, Glatz A, Nakamoto H, Mishkind ML, Munnik T, Saidi Y, Goloubinoff P, Harwood JL, Vigh L. Heat shock response in photosynthetic organisms: membrane and lipid connections. Prog Lipid Res 2012; 51:208-20. [PMID: 22484828 DOI: 10.1016/j.plipres.2012.02.002] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 01/31/2012] [Accepted: 02/01/2012] [Indexed: 11/29/2022]
Abstract
The ability of photosynthetic organisms to adapt to increases in environmental temperatures is becoming more important with climate change. Heat stress is known to induce heat-shock proteins (HSPs) many of which act as chaperones. Traditionally, it has been thought that protein denaturation acts as a trigger for HSP induction. However, increasing evidence has shown that many stress events cause HSP induction without commensurate protein denaturation. This has led to the membrane sensor hypothesis where the membrane's physical and structural properties play an initiating role in the heat shock response. In this review, we discuss heat-induced modulation of the membrane's physical state and changes to these properties which can be brought about by interaction with HSPs. Heat stress also leads to changes in lipid-based signaling cascades and alterations in calcium transport and availability. Such observations emphasize the importance of membranes and their lipids in the heat shock response and provide a new perspective for guiding further studies into the mechanisms that mediate cellular and organismal responses to heat stress.
Collapse
Affiliation(s)
- Ibolya Horváth
- Institute of Biochemistry, Biol. Res. Centre, Hungarian Acad. Sci., Temesvári krt. 62, H-6734 Szeged, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Schilling D, Bayer C, Li W, Molls M, Vaupel P, Multhoff G. Radiosensitization of normoxic and hypoxic h1339 lung tumor cells by heat shock protein 90 inhibition is independent of hypoxia inducible factor-1α. PLoS One 2012; 7:e31110. [PMID: 22347438 PMCID: PMC3274537 DOI: 10.1371/journal.pone.0031110] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 01/02/2012] [Indexed: 11/30/2022] Open
Abstract
Background Ionizing irradiation is a commonly accepted treatment modality for lung cancer patients. However, the clinical outcome is hampered by normal tissue toxicity and tumor hypoxia. Since tumors often have higher levels of active heat shock protein 90 (Hsp90) than normal tissues, targeting of Hsp90 might provide a promising strategy to sensitize tumors towards irradiation. Hsp90 client proteins include oncogenic signaling proteins, cell cycle activators, growth factor receptors and hypoxia inducible factor-1α (HIF-1α). Overexpression of HIF-1α is assumed to promote malignant transformation and tumor progression and thus might reduce the accessibility to radiotherapy. Methodology/Principal Findings Herein, we describe the effects of the novel Hsp90 inhibitor NVP-AUY922 and 17-allylamino-17-demethoxygeldanamycin (17-AAG), as a control, on HIF-1α levels and radiosensitivity of lung carcinoma cells under normoxic and hypoxic conditions. NVP-AUY922 exhibited a similar biological activity to that of 17-AAG, but at only 1/10 of the dose. As expected, both inhibitors reduced basal and hypoxia-induced HIF-1α levels in EPLC-272H lung carcinoma cells. However, despite a down-regulation of HIF-1α upon Hsp90 inhibition, sensitivity towards irradiation remained unaltered in EPLC-272H cells under normoxic and hypoxic conditions. In contrast, treatment of H1339 lung carcinoma cells with NVP-AUY922 and 17-AAG resulted in a significant up-regulation of their initially high HIF-1α levels and a concomitant increase in radiosensitivity. Conclusions/Significance In summary, our data show a HIF-1α-independent radiosensitization of normoxic and hypoxic H1339 lung cancer cells by Hsp90 inhibition.
Collapse
Affiliation(s)
- Daniela Schilling
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Clinical Cooperation Group (CCG) “Innate Immunity in Tumor Biology”, Helmholtz Zentrum München, Munich, Germany
| | - Christine Bayer
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Wei Li
- Department of Dermatology, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Michael Molls
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Peter Vaupel
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Clinical Cooperation Group (CCG) “Innate Immunity in Tumor Biology”, Helmholtz Zentrum München, Munich, Germany
- * E-mail:
| |
Collapse
|
91
|
Farwanah H, Kolter T. Lipidomics of glycosphingolipids. Metabolites 2012; 2:134-64. [PMID: 24957371 PMCID: PMC3901200 DOI: 10.3390/metabo2010134] [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: 11/28/2011] [Revised: 01/27/2012] [Accepted: 01/30/2012] [Indexed: 01/14/2023] Open
Abstract
Glycosphingolipids (GSLs) contain one or more sugars that are attached to a sphingolipid moiety, usually to a ceramide, but in rare cases also to a sphingoid base. A large structural heterogeneity results from differences in number, identity, linkage, and anomeric configuration of the carbohydrate residues, and also from structural differences within the hydrophobic part. GSLs form complex cell-type specific patterns, which change with the species, the cellular differentiation state, viral transformation, ontogenesis, and oncogenesis. Although GSL structures can be assigned to only a few series with a common carbohydrate core, their structural variety and the complex pattern are challenges for their elucidation and quantification by mass spectrometric techniques. We present a general overview of the application of lipidomics for GSL determination. This includes analytical procedures and instrumentation together with recent correlations of GSL molecular species with human diseases. Difficulties such as the structural complexity and the lack of standard substances for complex GSLs are discussed.
Collapse
Affiliation(s)
- Hany Farwanah
- Life and Medical Sciences Institute (LiMES), Membrane Biology and Lipid Biochemistry Unit, c/o Kekulé-Institut für Organische Chemie und Biochemie, University of Bonn, Gerhard-Domagk Str. 1, D-53121 Bonn, Germany.
| | - Thomas Kolter
- Life and Medical Sciences Institute (LiMES), Membrane Biology and Lipid Biochemistry Unit, c/o Kekulé-Institut für Organische Chemie und Biochemie, University of Bonn, Gerhard-Domagk Str. 1, D-53121 Bonn, Germany.
| |
Collapse
|
92
|
Multhoff G, Molls M, Radons J. Chronic inflammation in cancer development. Front Immunol 2012; 2:98. [PMID: 22566887 PMCID: PMC3342348 DOI: 10.3389/fimmu.2011.00098] [Citation(s) in RCA: 314] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 12/28/2011] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammatory mediators exert pleiotropic effects in the development of cancer. On the one hand, inflammation favors carcinogenesis, malignant transformation, tumor growth, invasion, and metastatic spread; on the other hand inflammation can stimulate immune effector mechanisms that might limit tumor growth. The link between cancer and inflammation depends on intrinsic and extrinsic pathways. Both pathways result in the activation of transcription factors such as NF-κB, STAT-3, and HIF-1 and in accumulation of tumorigenic factors in tumor and microenvironment. STAT-3 and NF-κB interact at multiple levels and thereby boost tumor-associated inflammation which can suppress anti-tumor immune responses. These factors also promote tumor growth, progression, and metastatic spread. IL-1, IL-6, TNF, and PGHS-2 are key mediators of an inflammatory milieu by modulating the expression of tumor-promoting factors. In this review we concentrate on the crucial role of pro-inflammatory mediators in inflammation-driven carcinogenesis and outline molecular mechanisms of IL-1 signaling in tumors. In addition, we elucidate the dual roles of stress proteins as danger signals in the development of anti-cancer immunity and anti-apoptotic functions.
Collapse
Affiliation(s)
- Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München Munich, Germany.
| | | | | |
Collapse
|
93
|
Boroughs LK, Antonyak MA, Johnson JL, Cerione RA. A unique role for heat shock protein 70 and its binding partner tissue transglutaminase in cancer cell migration. J Biol Chem 2011; 286:37094-107. [PMID: 21896482 PMCID: PMC3199457 DOI: 10.1074/jbc.m111.242438] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 08/02/2011] [Indexed: 01/23/2023] Open
Abstract
Cell migration is essential for several important biological outcomes and is involved in various developmental disorders and disease states including cancer cell invasiveness and metastasis. A fundamental step in cell migration is the development of a leading edge. By using HeLa carcinoma cells as an initial model system, we uncovered a surprising role for the heat shock protein 70 (Hsp70) and its ability to bind the protein cross-linking enzyme, tissue transglutaminase (tTG), in cancer cell migration. Treatment of HeLa cells with EGF results in the activation of a plasma membrane-associated pool of tTG and its redistribution to the leading edges of these cells, which are essential events for EGF-stimulated HeLa cell migration. However, we then found that the ability of tTG to be localized to the leading edge is dependent on Hsp70. Similarly, the localization of tTG to the leading edges of MDAMB231 breast carcinoma cells, where it also plays an essential role in their migration, has a strict requirement for Hsp70. Treatment of these different cell lines with inhibitors against the ATP hydrolytic activity of Hsp70 prevented tTG from localizing to their leading edges and thereby blocked EGF-stimulated HeLa cell migration, as well as the constitutive migration normally exhibited by MDAMB231 cells. These findings highlight a new and unconventional role for the chaperonin activity of Hsp70 in the localization of a key regulatory protein (tTG) at the leading edges of cancer cells and the important consequences that this holds for their ability to migrate.
Collapse
Affiliation(s)
- Lindsey K. Boroughs
- From the Department of Molecular Medicine, Cornell University, Ithaca, New York 14853
| | - Marc A. Antonyak
- From the Department of Molecular Medicine, Cornell University, Ithaca, New York 14853
| | - Jared L. Johnson
- From the Department of Molecular Medicine, Cornell University, Ithaca, New York 14853
| | - Richard A. Cerione
- From the Department of Molecular Medicine, Cornell University, Ithaca, New York 14853
| |
Collapse
|
94
|
Zhu YZ, Cao MM, Wang WB, Wang W, Ren H, Zhao P, Qi ZT. Association of heat-shock protein 70 with lipid rafts is required for Japanese encephalitis virus infection in Huh7 cells. J Gen Virol 2011; 93:61-71. [PMID: 21940409 DOI: 10.1099/vir.0.034637-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Japanese encephalitis virus (JEV) is an enveloped flavivirus and the most common agent of viral encephalitis. It enters cells through receptor-mediated endocytosis and low pH-triggered membrane fusion. Although lipid rafts, cholesterol-enriched lipid-ordered membrane domains, have been shown to participate in JEV entry, the mechanisms of the early events of JEV infection, including the cellular receptors of JEV, remain largely unknown. In the current study, it was demonstrated that heat-shock protein 70 (HSP70), rather than other members of the HSP70 family, was required for JEV entry into a human cell line. Cell-surface expression of HSP70 and a direct interaction between JEV envelope (E) protein and HSP70 were observed. Biochemical fractionation showed that HSP70 clearly migrated into the raft fraction after virus infection and co-fractioned with E protein. Depletion of cholesterol shifted the E protein and HSP70 to a non-raft membrane and decreased JEV entry without affecting virus binding to host cells. Notably, recruitment of HSP70 into lipid rafts was required for activation of the phosphoinositide 3-kinase/Akt signalling pathway in the early stage of JEV infection. These results indicate that lipid rafts facilitate JEV entry, possibly by providing a convenient platform to concentrate JEV and its receptors on the host-cell membrane.
Collapse
Affiliation(s)
- Yong-Zhe Zhu
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, PR China
| | - Ming-Mei Cao
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, PR China
| | - Wen-Bo Wang
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, PR China
| | - Wen Wang
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, PR China
| | - Hao Ren
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, PR China
| | - Ping Zhao
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, PR China
| | - Zhong-Tian Qi
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, PR China
| |
Collapse
|
95
|
Kepp O, Galluzzi L, Martins I, Schlemmer F, Adjemian S, Michaud M, Sukkurwala AQ, Menger L, Zitvogel L, Kroemer G. Molecular determinants of immunogenic cell death elicited by anticancer chemotherapy. Cancer Metastasis Rev 2011; 30:61-9. [PMID: 21249425 DOI: 10.1007/s10555-011-9273-4] [Citation(s) in RCA: 220] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The success of some chemo- and radiotherapeutic regimens relies on the induction of immunogenic tumor cell death and on the induction of an anticancer immune response. Cells succumbing to immunogenic cell death undergo specific changes in their surface characteristics and release pro-immunogenic factors according to a defined spatiotemporal pattern. This stimulates antigen presenting cells such as dendritic cells to efficiently take up tumor antigens, process them, and cross-prime cytotoxic T lymphocytes, thus eliciting a tumor-specific cognate immune response. Such a response can also target therapy-resistant tumor (stem) cells, thereby leading, at least in some instances, to tumor eradication. In this review, we shed some light on the molecular identity of the factors that are required for cell death to be perceived as immunogenic. We discuss the intriguing observations that the most abundant endoplasmic reticulum protein, calreticulin, the most abundant intracellular metabolite, ATP, and the most abundant non-histone chromatin-binding protein, HMGB1, can determine whether cell death is immunogenic as they appear on the surface or in the microenvironment of dying cells.
Collapse
Affiliation(s)
- Oliver Kepp
- INSERM, U848, Institut Gustave Roussy, Pavillon de Recherche 1, 94805 Villejuif (Paris), France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Stangl S, Gehrmann M, Dressel R, Alves F, Dullin C, Themelis G, Ntziachristos V, Staeblein E, Walch A, Winkelmann I, Multhoff G. In vivo imaging of CT26 mouse tumours by using cmHsp70.1 monoclonal antibody. J Cell Mol Med 2011; 15:874-87. [PMID: 20406322 PMCID: PMC3922674 DOI: 10.1111/j.1582-4934.2010.01067.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The major stress-inducible heat shock protein 70 (Hsp70) is frequently present on the cell surface of human tumours, but not on normal cells. Herein, the binding characteristics of the cmHsp70.1 mouse monoclonal antibody (mAb) were evaluated in vitro and in a syngeneic tumour mouse model. More than 50% of the CT26 mouse colon carcinoma cells express Hsp70 on their cell surface at 4°C. After a temperature shift to 37°C, the cmHsp70.1-fluorescein isothiocyanate mAb translocates into early endosomes and lysosomes. Intraoperative and near-infrared fluorescence imaging revealed an enrichment of Cy5.5-conjugated mAb cmHsp70.1, but not an identically labelled IgG1 isotype-matched control, in i.p. and s.c. located CT26 tumours, as soon as 30 min. after i.v. injection into the tail vein. Due to the rapid turnover rate of membrane-bound Hsp70, the fluorescence-labelled cmHsp70.1 mAb became endocytosed and accumulated in the tumour, reaching a maximum after 24 hrs and remained detectable at least up to 96 hrs after a single i.v. injection. The tumour-selective internalization of mAb cmHsp70.1 at the physiological temperature of 37°C might enable a targeted uptake of toxins or radionuclides into Hsp70 membrane-positive tumours. The anti-tumoral activity of the cmHsp70.1 mAb is further supported by its capacity to mediate antibody-dependent cytotoxicity.
Collapse
Affiliation(s)
- Stefan Stangl
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, and Clinical Cooperation Group (CCG) 'Innate Immunity in Tumor Biology', Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Donfack P, Multhoff G, Materny A. Label-free nondestructive discrimination of colon carcinoma sublines and biomolecular insights into their differential Hsp70 expression: DNA/RNA nucleobase specific changes. Chembiochem 2011; 12:1922-36. [PMID: 21739554 DOI: 10.1002/cbic.201000653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/19/2011] [Indexed: 11/11/2022]
Abstract
Hsp70 is biologically relevant for its chaperon functions. The CX(-) and CX(+) sublines, which derive from the parental colon carcinoma CX2 cell line, are accordingly very similar. They have been reported to be specifically different only in Hsp70 membrane expression, which is associated with immunostimulatory effects. CX(-) /CX(+) have been phenotypically characterized by immunofluorescence studies and Raman spectroscopy combined with robust clustering and multivariate analysis. With the latter we address the potential of overall characterization for CX(-) /CX(+) discrimination and gain molecular insights into Hsp70 differential expression. Due to their strong resemblance, CX(-) and CX(+) show similar mean Raman spectra, which look indiscernible at first. Interestingly, their rather protein-dominated Raman spectra reveal, besides changes in protein and amino acids, very specific changes in DNA/RNA nucleotides involving pyrimidine ring Raman hypochromic effects. Therefore, discriminating CX(-) from CX(+) is ultimately achieved based on principal component scores. Because CX(-) /CX(+) are associated with the same lipid marker, changes in proteins support lipid interactions with regulatory proteins. More importantly, changes observed in nucleobases, which are indicative of DNA/RNA-protein binding interactions, suggest transcription deregulations as participating precursor onsets of different transport mechanisms that lead to Hsp70 differential expression and associated phenotypic variation. Besides immunofluorescence, we have used Raman spectroscopy combined with multivariate analysis within an autologous tumor system for label-free nondestructive cell-subline discrimination, and demonstrate, to our knowledge, the first overall phenotypic monitoring with insights into Hsp70 differential expression. This might well prove to be useful for Raman label-free cell-sorting of the CX(-) /CX(+) sublines.
Collapse
Affiliation(s)
- Patrice Donfack
- Department of Physics, Molecular Life Science Center, Jacobs University, Campus Ring 1, 28759 Bremenm, Germany
| | | | | |
Collapse
|
98
|
Stangl S, Themelis G, Friedrich L, Ntziachristos V, Sarantopoulos A, Molls M, Skerra A, Multhoff G. Detection of irradiation-induced, membrane heat shock protein 70 (Hsp70) in mouse tumors using Hsp70 Fab fragment. Radiother Oncol 2011; 99:313-6. [DOI: 10.1016/j.radonc.2011.05.051] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 05/20/2011] [Accepted: 05/20/2011] [Indexed: 11/25/2022]
|
99
|
Kolter T. A view on sphingolipids and disease. Chem Phys Lipids 2011; 164:590-606. [PMID: 21570958 DOI: 10.1016/j.chemphyslip.2011.04.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 04/26/2011] [Accepted: 04/28/2011] [Indexed: 12/27/2022]
Abstract
Sphingolipid and glycosphingolipid levels and expression of sphingolipid metabolizing enzymes are altered in a variety of diseases or in response to drug treatment. Inherited defects of enzymes and other proteins required for the lysosomal degradation of these lipids lead to human sphingolipidoses. Also genetic defects that affect sphingolipid biosynthesis are known. Although the molecular details are often far from clear, (glyco)sphingolipids have been implicated to play a role in atherosclerosis, insulin resistance, cancer, and infections by pathogens. More general aspects of selected diseases are discussed.
Collapse
Affiliation(s)
- Thomas Kolter
- LiMES-Laboratory of Lipid Biochemistry, Kekulé-Institut für Organische Chemie und Biochemie der Universität, Bonn, Germany.
| |
Collapse
|
100
|
Multhoff G, Hightower LE. Distinguishing integral and receptor-bound heat shock protein 70 (Hsp70) on the cell surface by Hsp70-specific antibodies. Cell Stress Chaperones 2011; 16:251-5. [PMID: 21165727 PMCID: PMC3077224 DOI: 10.1007/s12192-010-0247-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 11/24/2010] [Indexed: 01/10/2023] Open
Abstract
Cell Stress & Chaperones journal has become a major outlet for papers and review articles about anti-heat shock protein (HSP) antibodies. In the last decade, it became evident that apart from their intracellular localization, members of the heat shock protein 90 (Hsp90; HSPC) and Hsp70 (HSPA) family are also found on the cell surface. In this review, we will focus on Hsp70 (HSPA1A), the major stress-inducible member of the human Hsp70 family. Depending on the cell type, the membrane association of Hsp70 comes in two forms. In tumor cells, Hsp70 appears to be integrated within the plasma membrane, whereas in non-malignantly transformed (herein termed normal) cells, Hsp70 is associated with cell surface receptors. This observation raises the question whether or not these two surface forms of Hsp70 in tumor and normal cells can be distinguished using Hsp70 specific antibodies. Presently a number of Hsp70 specific antibodies are commercially available. These antibodies were generated by immunizing mice either with recombinant or HeLa-derived human Hsp70 protein, parts of the Hsp70 protein, or with synthetic peptides. This review aims to characterize the binding of different anti-human Hsp70 antibodies and their capacity to distinguish between integrated and receptor-bound Hsp70 in tumor and normal cells.
Collapse
Affiliation(s)
- Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Ismaningerstrasse 22, Munich, Germany.
| | | |
Collapse
|