92 results on '"Lars H. Engelholm"'
Search Results
2. Supplementary Figure 6 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Author
-
Kaisa Lehti, Jorma Keski-Oja, Kari Alitalo, Jussi Taipale, Lars H. Engelholm, Signe Ingvarsen, Olli Kallioniemi, Sami Kilpinen, Marko Hyytiäinen, Jouko Lohi, Pipsa Meller, Markku Varjosalo, and Nami Sugiyama
- Abstract
Supplementary Figure 6 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figure 7 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Author
-
Kaisa Lehti, Jorma Keski-Oja, Kari Alitalo, Jussi Taipale, Lars H. Engelholm, Signe Ingvarsen, Olli Kallioniemi, Sami Kilpinen, Marko Hyytiäinen, Jouko Lohi, Pipsa Meller, Markku Varjosalo, and Nami Sugiyama
- Abstract
Supplementary Figure 7 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Legends for Figures 1-8, Tables 1-3, Methods from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Author
-
Kaisa Lehti, Jorma Keski-Oja, Kari Alitalo, Jussi Taipale, Lars H. Engelholm, Signe Ingvarsen, Olli Kallioniemi, Sami Kilpinen, Marko Hyytiäinen, Jouko Lohi, Pipsa Meller, Markku Varjosalo, and Nami Sugiyama
- Abstract
Supplementary Legends for Figures 1-8, Tables 1-3, Methods from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Tables 1-3 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Author
-
Kaisa Lehti, Jorma Keski-Oja, Kari Alitalo, Jussi Taipale, Lars H. Engelholm, Signe Ingvarsen, Olli Kallioniemi, Sami Kilpinen, Marko Hyytiäinen, Jouko Lohi, Pipsa Meller, Markku Varjosalo, and Nami Sugiyama
- Abstract
Supplementary Tables 1-3 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 5 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Author
-
Kaisa Lehti, Jorma Keski-Oja, Kari Alitalo, Jussi Taipale, Lars H. Engelholm, Signe Ingvarsen, Olli Kallioniemi, Sami Kilpinen, Marko Hyytiäinen, Jouko Lohi, Pipsa Meller, Markku Varjosalo, and Nami Sugiyama
- Abstract
Supplementary Figure 5 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 2 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Author
-
Kaisa Lehti, Jorma Keski-Oja, Kari Alitalo, Jussi Taipale, Lars H. Engelholm, Signe Ingvarsen, Olli Kallioniemi, Sami Kilpinen, Marko Hyytiäinen, Jouko Lohi, Pipsa Meller, Markku Varjosalo, and Nami Sugiyama
- Abstract
Supplementary Figure 2 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 3 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Author
-
Kaisa Lehti, Jorma Keski-Oja, Kari Alitalo, Jussi Taipale, Lars H. Engelholm, Signe Ingvarsen, Olli Kallioniemi, Sami Kilpinen, Marko Hyytiäinen, Jouko Lohi, Pipsa Meller, Markku Varjosalo, and Nami Sugiyama
- Abstract
Supplementary Figure 3 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 4 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Author
-
Kaisa Lehti, Jorma Keski-Oja, Kari Alitalo, Jussi Taipale, Lars H. Engelholm, Signe Ingvarsen, Olli Kallioniemi, Sami Kilpinen, Marko Hyytiäinen, Jouko Lohi, Pipsa Meller, Markku Varjosalo, and Nami Sugiyama
- Abstract
Supplementary Figure 4 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure 1 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Author
-
Kaisa Lehti, Jorma Keski-Oja, Kari Alitalo, Jussi Taipale, Lars H. Engelholm, Signe Ingvarsen, Olli Kallioniemi, Sami Kilpinen, Marko Hyytiäinen, Jouko Lohi, Pipsa Meller, Markku Varjosalo, and Nami Sugiyama
- Abstract
Supplementary Figure 1 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figures 1 - 6 from Tumor MMP-1 Activates Endothelial PAR1 to Facilitate Vascular Intravasation and Metastatic Dissemination
- Author
-
James P. Quigley, Lars H. Engelholm, Tatyana A. Kupriyanova, Ewa Zajac, Ivo Rimann, Elena I. Deryugina, and Anna Juncker-Jensen
- Abstract
PDF file - 199K, Effects of MMP-1 silencing on HEp3-hi/diss cell proliferation in 2D and 3D culture conditions (S1); HEp3-hi/diss tissue colonization does not depend on MMP-1 expression (S2); Effects of MMP-1 downregulation on HEp3-hi/diss cell adhesion and invasion (S3); Inhibition of vascular permeability in HEp3-hi/diss microtumors by the PAR1 antagonist RWJ 56110 (S4); Inhibition of HEp3-hi/diss intravasation by a PAR1 antagonist (S5); MMP-1 produced by the human HNSCC Detroit 562 concomitantly regulates vascular permeability and intravasation (S6).
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 8 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Author
-
Kaisa Lehti, Jorma Keski-Oja, Kari Alitalo, Jussi Taipale, Lars H. Engelholm, Signe Ingvarsen, Olli Kallioniemi, Sami Kilpinen, Marko Hyytiäinen, Jouko Lohi, Pipsa Meller, Markku Varjosalo, and Nami Sugiyama
- Abstract
Supplementary Figure 8 from Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Published
- 2023
- Full Text
- View/download PDF
13. Targeted delivery of alcohol-containing payloads with antibody-drug conjugates
- Author
-
Katja E. Grier, Anders H. Hansen, Christina S. Haxvig, Xin Li, Oliver Krigslund, Niels Behrendt, Lars H. Engelholm, Fabio Rossi, Bebiana C. Sousa, Grant J. Harradence, Nicolas Camper, and Katrine M. Qvortrup
- Subjects
Materials Chemistry ,Metals and Alloys ,Ceramics and Composites ,General Chemistry ,Catalysis ,Surfaces, Coatings and Films ,Electronic, Optical and Magnetic Materials - Abstract
We herein describe the cell-specific release of alcohol-containing payloads via a sulfatase-sensitive linker in antibody-drug conjugates (ADCs).
- Published
- 2023
- Full Text
- View/download PDF
14. Abstract 5779: A platform utilizing high-grade serous ovarian cancer organoids for prospective patient stratification in functional precision medicine
- Author
-
Wojciech Senkowski, Laura Gall-Mas, Matias M. Falco, Yilin Li, Kari Lavikka, Mette C. Kriegbaum, Jaana Oikkonen, Daria Bulanova, Elin J. Pietras, Karolin Voßgröne, Yan-Jun Chen, Erdogan P. Erkan, Mia K. Høg, Ida M. Larsen, Tarja Lamminen, Katja Kaipio, Jutta Huvila, Anni Virtanen, Lars H. Engelholm, Pernille Christiansen, Eric Santoni Rugiu, Kaisa Huhtinen, Olli Carpén, Johanna Hynninen, Sampsa Hautaniemi, Anna Vähärautio, and Krister Wennerberg
- Subjects
Cancer Research ,Oncology - Abstract
High-grade serous ovarian cancer (HGSC) is the most prevalent and lethal ovarian cancer type. While HGSC usually responds well to primary treatment, most cases eventually relapse. Functional precision medicine - tailoring individualized treatments based on functional in vitro assays on patient-derived cells - has been recently employed in cancer clinical trials. Cancer organoids - three-dimensional, self-organizing, self-renewing cell cultures that recapitulate original tissue structure and function - have been applied as cellular models in these trials. However, in case of HGSC, organoid derivation has proven time consuming and inefficient, hindering their application in functional precision medicine due to a short time window, in which therapy for each patient needs to be selected. To address this problem, we aimed to establish whether drug vulnerabilities at HGSC relapse could be predicted using organoids derived from the primary disease cells. We derived sequential organoid models from material sampled during primary treatment and at relapse. Then, for organoid pairs (primary-relapse), we performed large-scale drug response profiling of a library of 370 compounds (approved drugs or drugs in clinical development), in 384-well microplate format, alone or in combination with a standard HGSC chemotherapeutic agent carboplatin. First, we found that HGSC organoid responses to standard chemotherapeutics retrospectively correlated to observed clinical treatment outcomes. But further, for each patient we identified compounds with pronounced cytotoxicity both in the primary and in the relapsed model, amounting to 66% of all hits (7% were primary-specific and 27% relapse-specific). We then focused on identifying patient-specific hits rather than compounds displaying general toxicity in all patient models. Based on a potential clinical applicability, for three patients we selected compounds for validation in organoid outgrowth assay, with prolonged (>1 month) drug-free period post-treatment. In two patients, AZD4573, a selective CDK9 inhibitor in clinical development for hematological malignancies, at nanomolar concentrations caused eradication of organoids when combined with carboplatin. Organoids from the third patient were vulnerable to nitazoxanide, an approved anti-helminthic agent and an inhibitor of mitochondrial oxidative phosphorylation. Importantly, the selected final hits were identified solely based on screening in organoid models from primary disease. In summary, we here demonstrate that HGSC organoids derived from primary disease material predict a majority of patient-specific drug vulnerabilities of organoids derived from the relapsed HGSC lesions. This indicates that patient stratification in functional precision medicine for treatment of HGSC relapse could be prospectively performed at the primary disease stage. Citation Format: Wojciech Senkowski, Laura Gall-Mas, Matias M. Falco, Yilin Li, Kari Lavikka, Mette C. Kriegbaum, Jaana Oikkonen, Daria Bulanova, Elin J. Pietras, Karolin Voßgröne, Yan-Jun Chen, Erdogan P. Erkan, Mia K. Høg, Ida M. Larsen, Tarja Lamminen, Katja Kaipio, Jutta Huvila, Anni Virtanen, Lars H. Engelholm, Pernille Christiansen, Eric Santoni Rugiu, Kaisa Huhtinen, Olli Carpén, Johanna Hynninen, Sampsa Hautaniemi, Anna Vähärautio, Krister Wennerberg. A platform utilizing high-grade serous ovarian cancer organoids for prospective patient stratification in functional precision medicine. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5779.
- Published
- 2023
- Full Text
- View/download PDF
15. Abstract 1547: The urokinase plasminogen activator receptor-associated protein (uPARAP) is an attractive target for the development of antibody-drug conjugates (ADCs) for treatment of mesenchymal malignancies
- Author
-
Pernille Barkholt, Agnieszka Wozniak, Chao-Chi Wang, Che-Jui Lee, Lore De Kock, Lars H. Engelholm, Carmel Lynch, Dominik Mumberg, and Patrick Schöffski
- Subjects
Cancer Research ,Oncology - Abstract
Background: Soft tissue sarcoma (STS) is a complex family of rare malignancies with high unmet medical need. uPARAP is an endocytic receptor expressed at low levels on selected mesenchymal cell types, which internalizes fragments of collagen and transfers them to the lysosome for degradation. Besides its physiological role, uPARAP is considered a critical modulator of the tumor microenvironment. Here, for the first time, we explored the molecular epidemiology of uPARAP in sarcoma tissue to validate this receptor as target for the development of ADCs in this indication. Methods: uPARAP expression was assessed in STS-specific tissue microarrays by immunohistochemistry in tissue samples from more than three hundred individual donors. Stainings were assessed by histopathologic scoring and grouped into high, medium and low expressing subgroups. Results: uPARAP was found to be strongly over-expressed in a high percentage of cases of common STS, with variations in terms of incidence and level of expression between histological subtypes. High uPARAP expression was found in fibro- (86% of cases), synovial (84%), dedifferentiated lipo- (68%), pleomorphic lipo- (65%), myxofibro- (59%), leiomyo- (49%), and myxoid liposarcoma (24%). Work is ongoing in additional sarcoma subtypes. Conclusions: Based on protein expression uPARAP is an attractive emerging therapeutic target for the development of uPARAP-binding ADCs in a broad range of mesenchymal malignancies. In addition, expression of uPARAP in STS may serve as a potential marker for patient selection in early clinical studies with uPARAP-targeting ADCs. Citation Format: Pernille Barkholt, Agnieszka Wozniak, Chao-Chi Wang, Che-Jui Lee, Lore De Kock, Lars H. Engelholm, Carmel Lynch, Dominik Mumberg, Patrick Schöffski. The urokinase plasminogen activator receptor-associated protein (uPARAP) is an attractive target for the development of antibody-drug conjugates (ADCs) for treatment of mesenchymal malignancies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 1547.
- Published
- 2023
- Full Text
- View/download PDF
16. The collagen receptor uPARAP/Endo180 regulates collectins through unique structural elements in its FNII domain
- Author
-
Niels Behrendt, Lars H. Engelholm, Kirstine S Nørregaard, Henrik J. Jürgensen, and Oliver Krigslund
- Subjects
0301 basic medicine ,Receptors, Collagen ,Endocytic cycle ,Collectin ,Receptors, Cell Surface ,CHO Cells ,Mannose-Binding Lectin ,Biochemistry ,Receptors, Urokinase Plasminogen Activator ,Collagen receptor ,03 medical and health sciences ,Cricetulus ,Animals ,Humans ,Lectins, C-Type ,Molecular Biology ,Mannan-binding lectin ,Membrane Glycoproteins ,Innate immune system ,030102 biochemistry & molecular biology ,biology ,Chemistry ,Surfactant protein D ,Cell Biology ,Fibroblasts ,Pulmonary Surfactant-Associated Protein D ,Collectins ,Endocytosis ,Cell biology ,Fibronectin ,HEK293 Cells ,Mannose-Binding Lectins ,030104 developmental biology ,Receptors, Mitogen ,biology.protein ,Collagen ,Carrier Proteins ,Mannose Receptor ,Mannose receptor - Abstract
C-type lectins that contain collagen-like domains are known as collectins. These proteins are present both in the circulation and in extravascular compartments and are central players of the innate immune system, contributing to first-line defenses against viral, bacterial, and fungal pathogens. The collectins mannose-binding lectin (MBL) and surfactant protein D (SP-D) are regulated by tissue fibroblasts at extravascular sites via an endocytic mechanism governed by urokinase plasminogen activator receptor–associated protein (uPARAP or Endo180), which is also a collagen receptor. Here, we investigated the molecular mechanisms that drive the uPARAP-mediated cellular uptake of MBL and SP-D. We found that the uptake depends on residues within a protruding loop in the fibronectin type-II (FNII) domain of uPARAP that are also critical for collagen uptake. Importantly, however, we also identified FNII domain residues having an exclusive role in collectin uptake. We noted that these residues are absent in the related collagen receptor, the mannose receptor (MR or CD206), which consistently does not interact with collectins. We also show that the second C-type lectin-like domain (CTLD2) is critical for the uptake of SP-D, but not MBL, indicating an additional level of complexity in the interactions between collectins and uPARAP. Finally, we demonstrate that the same molecular mechanisms enable uPARAP to engage MBL immobilized on the surface of pathogens, thereby expanding the potential biological implications of this interaction. Our study reveals molecular details of the receptor-mediated cellular regulation of collectins and offers critical clues for future investigations into collectin biology and pathology.
- Published
- 2020
- Full Text
- View/download PDF
17. The endocytic receptor uPARAP is a regulator of extracellular thrombospondin-1
- Author
-
Kirstine S. Nørregaard, Henrik J. Jürgensen, Signe Z. Ingvarsen, Signe S. Heltberg, Christina E. Hagensen, Henrik Gårdsvoll, Daniel H. Madsen, Ole N. Jensen, Lars H. Engelholm, and Niels Behrendt
- Subjects
Mice, Knockout ,Proteomics ,Mannose receptor protein family ,Membrane Glycoproteins ,extracellular matrix ,Receptors, Cell Surface ,Ligands ,Endocytosis ,Thrombospondin 1 ,Mice ,proteomics ,endocytosis ,Animals ,Collagen ,Molecular Biology ,uPARAP/Endo180 ,mass spectrometry - Abstract
Thrombospondin-1 (TSP-1) is a matricellular protein with a multitude of functions in the pericellular and extracellular environment. We report a novel pathway for the regulation of extracellular TSP-1, governed by the endocytic collagen receptor, uPARAP (urokinase plasminogen activator receptor-associated protein; MRC2 gene product, also designated Endo180, CD280). First, using a novel proteomic approach for unbiased identification of ligands for endocytosis, we identify TSP-1 as a candidate ligand for specific uptake by uPARAP. We then show that uPARAP can efficiently internalize TSP-1 for lysosomal degradation, that this capability is not shared by other, closely related endocytic receptors and that uPARAP serves to regulate the extracellular levels of TSP-1 in vitro. Using wild type and uPARAP null mice, we also demonstrate uPARAP-mediated endocytosis of TSP-1 in dermal fibroblasts in vivo. Unlike other uPARAP ligands, the interaction with TSP-1 is sensitive to heparin and the responsible molecular motifs in uPARAP are overlapping, but not identical with those governing the interaction with collagens. Finally, we show that uPARAP can also mediate the endocytosis of TSP-2, a thrombospondin closely related to TSP-1, but not the more distantly related members of the same protein family, TSP-3, -4 and -5. These findings indicate that the role of uPARAP in ECM remodeling is not limited to the uptake of collagen for degradation but also includes an orchestrator function in the regulation of thrombospondins with numerous downstream effects. This is likely to be an important factor in the physiological and pathological roles of uPARAP in bone biology, fibrosis and cancer. The proteomic data has been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier PXD031272.
- Published
- 2022
- Full Text
- View/download PDF
18. Uncovering mediators of collagen degradation in the tumor microenvironment
- Author
-
Marie-Louise Thorseth, Marco Carretta, Christina Jensen, Kasper Mølgaard, Henrik J. Jürgensen, Lars H. Engelholm, Niels Behrendt, Nicholas Willumsen, and Daniel H. Madsen
- Subjects
Collagen endocytosis ,Histology ,QH301-705.5 ,Tumor-associated macrophages ,Biophysics ,Cell Biology ,Biochemistry ,Collagen degradation ,Extracellular matrix remodeling ,Matrix metalloproteinases ,Tumor microenvironment ,Genetics ,Biology (General) ,Molecular Biology ,Cancer-associated fibroblasts - Abstract
Increased remodeling of the extracellular matrix in malignant tumors has been shown to correlate with tumor aggressiveness and a poor prognosis. This remodeling involves degradation of the original extracellular matrix (ECM) and deposition of a new tumor-supporting ECM. The main constituent of the ECM is collagen and collagen turnover mainly occurs in a sequential manner, where initial proteolytic cleavage of the insoluble fibers is followed by cellular internalization of large well-defined collagen fragments for lysosomal degradation. However, despite extensive research in the field, a lack of consensus on which cell types within the tumor microenvironment express the involved proteases still exists. Furthermore, the relative contribution of different cell types to collagen internalization is not well-established. Here, we developed quantitative ex vivo collagen degradation assays and show that the proteases responsible for the initial collagen cleavage in two murine syngeneic tumor models are matrix metalloproteinases produced by cancer-associated fibroblasts and that collagen degradation fragments are endocytosed primarily by tumor-associated macrophages and cancer-associated fibroblasts from the tumor stroma. Using tumors from mannose receptor-deficient mice, we show that this receptor is essential for collagen-internalization by tumor-associated macrophages. Together, these findings identify the cell types responsible for the entire collagen degradation pathway, from initial cleavage to endocytosis of fragments for intracellular degradation.
- Published
- 2022
- Full Text
- View/download PDF
19. The Urokinase Receptor (uPAR) as a 'Trojan Horse' in Targeted Cancer Therapy: Challenges and Opportunities
- Author
-
Michael Ploug, Virginia Metrangolo, and Lars H. Engelholm
- Subjects
Cytotoxic approaches ,theranostics ,Cancer Research ,medicine.medical_treatment ,Cell ,Context (language use) ,Translational research ,Review ,urokinase plasminogen activator receptor (uPAR) ,Targeted therapy ,Extracellular matrix ,Medicine ,RC254-282 ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,cytotoxic approaches ,Theranostics ,targeted therapy ,Urokinase plasminogen activator receptor (uPAR) ,Urokinase receptor ,medicine.anatomical_structure ,translational research ,Oncology ,Cancer research ,Signal transduction ,business ,Plasminogen activator - Abstract
Simple Summary Discovered more than three decades ago, the urokinase-type plasminogen activator receptor (uPAR) has now firmly established itself as a versatile molecular target holding promise for the treatment of aggressive malignancies. The copious abundance of uPAR in virtually all human cancerous tissues versus their healthy counterparts has fostered a gradual shift in the therapeutic landscape targeting this receptor from function inhibition to cytotoxic approaches to selectively eradicate the uPAR-expressing cells by delivering a targeted cytotoxic insult. Multiple avenues are being explored in a preclinical setting, including the more innovative immune- or stroma targeting therapies. This review discusses the current state of these strategies, their potentialities, and challenges, along with future directions in the field of uPAR targeting. Abstract One of the largest challenges to the implementation of precision oncology is identifying and validating selective tumor-driving targets to enhance the therapeutic efficacy while limiting off-target toxicity. In this context, the urokinase-type plasminogen activator receptor (uPAR) has progressively emerged as a promising therapeutic target in the management of aggressive malignancies. By focalizing the plasminogen activation cascade and subsequent extracellular proteolysis on the cell surface of migrating cells, uPAR endows malignant cells with a high proteolytic and migratory potential to dissolve the restraining extracellular matrix (ECM) barriers and metastasize to distant sites. uPAR is also assumed to choreograph multiple other neoplastic stages via a complex molecular interplay with distinct cancer-associated signaling pathways. Accordingly, high uPAR expression is observed in virtually all human cancers and is frequently associated with poor patient prognosis and survival. The promising therapeutic potential unveiled by the pleiotropic nature of this receptor has prompted the development of distinct targeted intervention strategies. The present review will focus on recently emerged cytotoxic approaches emphasizing the novel technologies and related limits hindering their application in the clinical setting. Finally, future research directions and emerging opportunities in the field of uPAR targeting are also discussed.
- Published
- 2021
- Full Text
- View/download PDF
20. The Collagen Receptor uPARAP in Malignant Mesothelioma: A Potential Diagnostic Marker and Therapeutic Target
- Author
-
Lars H. Engelholm, Eric Santoni-Rugiu, Henrik Gårdsvoll, Oliver Krigslund, Rikke Raagaard Sørensen, Christoffer Nielsen, Pınar Çakılkaya, Henrik J. Jürgensen, and Niels Behrendt
- Subjects
Mesothelioma ,Male ,Immunoconjugates ,Gene Expression ,Antibody‐drug conjugate ,MRC2 ,Collagen receptor ,0302 clinical medicine ,Medicine ,Biology (General) ,Spectroscopy ,0303 health sciences ,Tissue microarray ,Membrane Glycoproteins ,medicine.diagnostic_test ,Endo180 ,Extracellular matrix ,General Medicine ,Middle Aged ,Immunohistochemistry ,3. Good health ,Computer Science Applications ,Up-Regulation ,Chemistry ,Tumor microenvironment ,mesothelioma ,030220 oncology & carcinogenesis ,immunohistochemistry ,Female ,Adult ,Receptors, Collagen ,QH301-705.5 ,medicine.drug_class ,extracellular matrix ,Receptors, Cell Surface ,Monoclonal antibody ,Article ,Catalysis ,antibody-drug conjugate ,Flow cytometry ,Inorganic Chemistry ,03 medical and health sciences ,Downregulation and upregulation ,Cell Line, Tumor ,Biomarkers, Tumor ,tumor microenvironment ,Humans ,Physical and Theoretical Chemistry ,QD1-999 ,Molecular Biology ,030304 developmental biology ,Aged ,uPARAP ,business.industry ,Organic Chemistry ,Mesothelioma, Malignant ,UPARAP ,Mannose-Binding Lectins ,ADC ,Cell culture ,Receptors, Mitogen ,Cancer research ,business ,Transcriptome ,CD280 - Abstract
Malignant mesothelioma (MM) is a highly aggressive cancer with limited therapeutic options. We have previously shown that the endocytic collagen receptor, uPARAP, is upregulated in certain cancers and can be therapeutically targeted. Public RNA expression data display uPARAP overexpression in MM. Thus, to evaluate its potential use in diagnostics and therapy, we quantified uPARAP expression by immunohistochemical H-score in formalin-fixed paraffin-embedded bioptic/surgical human tissue samples and tissue microarrays. We detected pronounced upregulation of uPARAP in the three main MM subtypes compared to non-malignant reactive mesothelial proliferations, with higher expression in sarcomatoid and biphasic than in epithelioid MM. The upregulation appeared to be independent of patients’ asbestos exposure and unaffected after chemotherapy. Using immunoblotting, we demonstrated high expression of uPARAP in MM cell lines and no expression in a non-malignant mesothelial cell line. Moreover, we showed the specific internalization of an anti-uPARAP monoclonal antibody by the MM cell lines using flow cytometry-based assays and confocal microscopy. Finally, we demonstrated the sensitivity of these cells towards sub-nanomolar concentrations of an antibody-drug conjugate formed with the uPARAP-directed antibody and a potent cytotoxin that led to efficient, uPARAP-specific eradication of the MM cells. Further studies on patient cohorts and functional preclinical models will fully reveal whether uPARAP could be exploited in diagnostics and therapeutic targeting of MM.
- Published
- 2021
- Full Text
- View/download PDF
21. Propranolol reduces sarcoma growth and enhances the response to anti-CTLA4 therapy by modulating the tumor microenvironment
- Author
-
Lars H. Engelholm, Niels Junker, Marco Carretta, Lars Grøntved, Anne Mette Askehøj Rømer, Daniel H. Madsen, Astrid Z. Johansen, Klaire Yixin Fjæstad, Victor Goitea, and Marie-Louise Thorseth
- Subjects
Tumor microenvironment ,medicine.drug_class ,business.industry ,Angiogenesis ,T cell ,Propranolol ,medicine.disease ,medicine.anatomical_structure ,Immune system ,Tumor progression ,medicine ,Cancer research ,Fibrosarcoma ,business ,Beta blocker ,medicine.drug - Abstract
The nonselective beta blocker, propranolol, which for decades has been prescribed for treatment of cardiovascular conditions, has recently been used successfully to treat metastatic angiosarcoma. These results have led to an orphan drug designation by the European Medicines Agency for the treatment of soft tissue sarcomas. The anti-tumor effects of propranolol are suggested to involve the reduction of cancer cell proliferation as well as angiogenesis.Here, we have investigated the anti-angiogenic properties of propranolol in the context of stimulating an anti-tumor immune response. We show that oral administration of propranolol delays tumor progression of MCA205 fibrosarcoma tumors and increases the survival rate of tumor bearing mice. Propranolol works by reducing tumor angiogenesis and facilitating an anti-tumoral microenvironment with increased T cell infiltration and reduced infiltration of myeloid-derived suppressor cells (MDSCs). Using T cell deficient mice, we demonstrate that the full anti-tumor effect of propranolol requires the presence of T cells. Flow cytometry-based analysis and RNA sequencing of FACS-sorted cells show that propranolol-treatment leads to an upregulation of PD-L1 on tumor-associated macrophages (TAMs) and changes in their chemokine expression profile. Lastly, we observe that the co-administration of propranolol significantly enhances the efficacy of anti-CTLA4 therapy.Our results identify propranolol as an immune modulating agent, which can improve immune checkpoint inhibitor therapies in soft tissue sarcoma patients and potentially in other cancers.
- Published
- 2021
- Full Text
- View/download PDF
22. Osteosarcoma and metastasis associated bone degradation—a tale of osteoclast and malignant cell cooperativity
- Author
-
Lars H. Engelholm, Henrik J. Jürgensen, Kent Søe, Kirstine S Nørregaard, Niels Behrendt, and Henrik Gårdsvoll
- Subjects
Bone degradation ,QH301-705.5 ,Osteoclasts ,Bone Neoplasms ,Cell Communication ,Review ,Catalysis ,Bone resorption ,Bone remodeling ,Metastasis ,Inorganic Chemistry ,Osteoclast ,Osteogenesis ,osteosarcoma ,medicine ,Animals ,Humans ,Physical and Theoretical Chemistry ,Bone Resorption ,Biology (General) ,Molecular Biology ,QD1-999 ,Spectroscopy ,bone metastasis ,Osteosarcoma ,business.industry ,Organic Chemistry ,Bone metastasis ,Cancer ,vicious cycle ,Vicious cycle ,General Medicine ,medicine.disease ,Computer Science Applications ,Chemistry ,Primary bone ,medicine.anatomical_structure ,osteoclast ,Cancer research ,Disease Progression ,Bone Remodeling ,Disease Susceptibility ,business ,bone degradation - Abstract
Cancer-induced bone degradation is part of the pathological process associated with both primary bone cancers, such as osteosarcoma, and bone metastases originating from, e.g., breast, prostate, and colon carcinomas. Typically, this includes a cancer-dependent hijacking of processes also occurring during physiological bone remodeling, including osteoclast-mediated disruption of the inorganic bone component and collagenolysis. Extensive research has revealed the significance of osteoclast-mediated bone resorption throughout the course of disease for both primary and secondary bone cancer. Nevertheless, cancer cells representing both primary bone cancer and bone metastasis have also been implicated directly in bone degradation. We will present and discuss observations on the contribution of osteoclasts and cancer cells in cancer-associated bone degradation and reciprocal modulatory actions between these cells. The focus of this review is osteosarcoma, but we will also include relevant observations from studies of bone metastasis. Additionally, we propose a model for cancer-associated bone degradation that involves a collaboration between osteoclasts and cancer cells and in which both cell types may directly participate in the degradation process.
- Published
- 2021
- Full Text
- View/download PDF
23. Abstract 2016: Preclinical evaluation of uPARAP (MRC2) antibody-drug conjugates (ADCE-003,010,011) in osteosarcoma pdx models
- Author
-
Yifei Wang, Wendong Zhang, Zhongting Zhang, Xiangjun Tian, Rossana Lazcano, Pooja Hingorani, Michael Roth, Jonathan Gill, Douglas Harrison, Zhaohui Xu, Jing Wang, Niels Behrendt, Christoffer F. Nielsen, Lars H. Engelholm, and Richard Gorlick
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Antibody-drug Conjugates (ADCs) targeting differentially expressed tumor cell-surface antigens show robust clinical activity in several solid tumor cancers, but none is yet available for osteosarcoma. Due to the high unmet medical need in this indication, there is an urgent requirement to identify respective osteosarcoma cell-surface antigens and evaluate the antitumor activity of an ADC that can deliver tailored cytotoxic payloads to osteosarcoma tumors expressing these targets. Methods: We used an integrated proteomic and transcriptomic surfaceome profiling approach to identify cell-surface proteins that are highly expressed in osteosarcoma but minimally expressed on normal tissues. uPARAP (MRC2) was found to be enriched in osteosarcoma at both protein and mRNA levels. The cell-surface expression of uPARAP (MRC2) was further validated by IHC and flow cytometry with osteosarcoma cell lines, PDX models, and patient tumor tissue microarray. As a proof of concept, three MRC2 targeted ADCs with different Microtubulin- and alkylating agent types of payloads were tested in 8 osteosarcoma PDX models. uPARAP (MRC2) ADC, control ADC, or vehicle control was administered IV to mice harboring flank tumors at a dose of 3mg/kg, weekly X 3 (ADCE-003), 2mg/kg on Day 1 (ADCE-010), and 10mg/kg weekly X 2 (ADCE-011). EFS for treatment (T) and control (C) groups, minimum relative tumor volume (minRTV), and objective response measures were analyzed. Results: Western blotting confirmed the expression of uPARAP (MRC2) in 8 osteosarcoma cell lines and 8 PDX models. Flow cytometry further validated cell-surface localization and expression levels of uPARAP (MRC2) in 7 osteosarcoma cell lines. We then performed immunohistochemistry (IHC) staining using an osteosarcoma tissue microarray from 37 patients and 19 PDX models. uPARAP (MRC2) was expressed in 97% of the patient samples and 95% of the PDXs. 51% of the patient samples and 39% of the PDXs had an overall H-score of 100 or higher. Mice tolerated ADCs well with minimal toxicity. All 3 ADCs significantly prolonged EFS in 6/8 osteosarcoma models. Complete response (CR) or maintained CR were observed in 2 models in ADCE-010 and ADCE-011 groups, respectively. Conclusions: uPARAP (MRC2) is highly expressed in most osteosarcoma samples, which makes it a viable target for respective antigen targeting ADC therapies. Three uPARAP (MRC2) targeting ADCs showed antitumor activities in osteosarcoma preclinical models which warrant further investigation of uPARAP targeting ADC therapies for osteosarcoma. Citation Format: Yifei Wang, Wendong Zhang, Zhongting Zhang, Xiangjun Tian, Rossana Lazcano, Pooja Hingorani, Michael Roth, Jonathan Gill, Douglas Harrison, Zhaohui Xu, Jing Wang, Niels Behrendt, Christoffer F. Nielsen, Lars H. Engelholm, Richard Gorlick. Preclinical evaluation of uPARAP (MRC2) antibody-drug conjugates (ADCE-003,010,011) in osteosarcoma pdx models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2016.
- Published
- 2022
- Full Text
- View/download PDF
24. Abstract 3069: Efficient establishment and utilization of a high-grade serous ovarian cancer organoid biobank
- Author
-
Wojciech Senkowski, Laura Gall Mas, Matias M. Falco, Yilin Li, Kari Lavikka, Mette C. Kriegbaum, Jaana Oikkonen, Daria Bulanova, Elin J. Pietras, Karolin Voßgröne, Erdogan P. Erkan, Terese K. Høj, Mia K. Høg, Tarja Lamminen, Katja Kaipio, Anni Virtanen, Lars H. Engelholm, Pernille Christiansen, Eric Santoni-Rugiu, Kaisa Huhtinen, Olli Carpén, Johanna Hynninen, Sampsa Hautaniemi, Anna Vähärautio, and Krister Wennerberg
- Subjects
Cancer Research ,Oncology - Abstract
Extensive utilization of organoids from high-grade serous ovarian carcinoma (HGSOC), the most common and lethal ovarian cancer, has been hampered by low success rates of long-term culture and scarcity of fresh tumor material. Here we present the development of a novel method for efficient generation, expansion and use of HGSOC organoids from cryopreserved tumor material. First, we assessed commonly used organoid media components and found that supplements such as FGF-2, R-Spondin1, Wnt or Noggin had negative impact on the HGSOC organoid derivation. But further, we found that supplementation with FGF-4, which has not been used in cancer organoid culture before, is beneficial for HGSOC organoid growth. Through extensive testing of various supplements and their combinations, we designed two novel HGSOC organoid media formulations - Medium 1 (M1) and Medium 2 (M2). Using M1 and M2 enabled generation and long-term expansion of living HGSOC organoid biobank with markedly improved success rate than in previous reports (55% vs. 23-38%). The organoids were established from cryopreserved tumor material, demonstrating the feasibility of using frozen tissue biobanks for HGSOC organoid derivation. Overall, we generated a collection of 18 expandable HGSOC organoid lines from 11 patients, encompassing samples from different tissue sites and disease progression stages. We validated the organoids using whole-genome sequencing, immunohistochemistry and single-cell RNA sequencing and demonstrated that they are genetically and phenotypically representative of original patient samples over long-term culture. Based on available patient consents, we deposited 3 organoid lines in a publicly accessible biobank. Finally, we investigated whether organoid drug responses correlate to those observed earlier in the clinic in the corresponding patients. Organoid-based drug-response profiling of clinically used HGSOC drug collection was performed in 384-well microplate format. To explore whether growth conditions impact correlation between organoid drug responses and clinical response, we compared the organoid drug responses in the nutrient-rich M1/M2 growth media to the ones observed in human plasma-like medium (HPLM), supplemented with relevant niche factors from M1/M2. Organoid drug responses correlated with clinical treatment outcomes, but only for organoids maintained in HPLM (Spearman r = 0.987, p=0.007 in HPLM vs 0.607, p=0.167 in growth medium, n=7), highlighting the importance of culture conditions in organoid-based functional assays. Taken together, we introduce a resource for efficient development and use of HGSOC organoids from cryopreserved material in ovarian cancer research. Citation Format: Wojciech Senkowski, Laura Gall Mas, Matias M. Falco, Yilin Li, Kari Lavikka, Mette C. Kriegbaum, Jaana Oikkonen, Daria Bulanova, Elin J. Pietras, Karolin Voßgröne, Erdogan P. Erkan, Terese K. Høj, Mia K. Høg, Tarja Lamminen, Katja Kaipio, Anni Virtanen, Lars H. Engelholm, Pernille Christiansen, Eric Santoni-Rugiu, Kaisa Huhtinen, Olli Carpén, Johanna Hynninen, Sampsa Hautaniemi, Anna Vähärautio, Krister Wennerberg. Efficient establishment and utilization of a high-grade serous ovarian cancer organoid biobank [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3069.
- Published
- 2022
- Full Text
- View/download PDF
25. Tumor cell MT1-MMP is dispensable for osteosarcoma tumor growth, bone degradation and lung metastasis
- Author
-
Niels Behrendt, Andreas Kjaer, Carsten H. Nielsen, Henrik J. Jürgensen, Maria C. Melander, Signe Ingvarsen, Lars H. Engelholm, Collin Tran, Oliver Krigslund, Josephine A Meilstrup, Henrik Gårdsvoll, Sander van Putten, Thomas H. Bugge, and Kirstine S Nørregaard
- Subjects
EXPRESSION ,musculoskeletal diseases ,Lung Neoplasms ,Molecular biology ,Angiogenesis ,INVASION ,lcsh:Medicine ,Bone Neoplasms ,macromolecular substances ,Biology ,Article ,Bone and Bones ,Metastasis ,Extracellular matrix ,Mice ,DEFICIENT ,stomatognathic system ,POOR-PROGNOSIS ,Cell Line, Tumor ,Matrix Metalloproteinase 14 ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,lcsh:Science ,Cancer ,Cell Proliferation ,Osteosarcoma ,Multidisciplinary ,RECEPTOR ,lcsh:R ,PATHWAYS ,medicine.disease ,CANCER ,Primary tumor ,Primary bone ,Tumor progression ,Gene Knockdown Techniques ,ORTHOTOPIC MODEL ,Disease Progression ,Cancer research ,lcsh:Q ,CRISPR-Cas Systems ,COLLAGEN DEGRADATION - Abstract
The membrane-anchored matrix metalloprotease MT1-MMP is a potent collagenolytic enzyme with a well-established role in extracellular matrix turnover and cellular invasion into collagen-rich tissues. MT1-MMP is highly expressed in various types of cancer and has been demonstrated to be directly involved in several stages of tumor progression, including primary tumor growth, angiogenesis, invasion and metastasis. Osteosarcoma is the most common type of primary bone cancer. This disease is characterized by invasive tumor growth, leading to extensive bone destruction, and metastasis to the lungs. The tumor cells in human osteosarcoma display a strong expression of MT1-MMP, but the role of MT1-MMP in osteosarcoma progression is currently unknown. In this study, we investigated the role of MT1-MMP during various stages of osteosarcoma development. We utilized an optimized orthotopic murine osteosarcoma model and human osteosarcoma cells in which the MT1-MMP gene was knocked out using CRISPR/Cas9. We observed a strong expression of MT1-MMP in wildtype cells of both primary tumors and lung metastases, but, surprisingly, MT1-MMP deficiency did not affect primary tumor growth, bone degradation or the formation and growth of lung metastases. We therefore propose that, unlike findings reported in other cancers, tumor-expressed MT1-MMP is dispensable for all stages of osteosarcoma progression.
- Published
- 2020
- Full Text
- View/download PDF
26. Personalized B cell response to the Lactobacillus rhamnosus GG probiotic in healthy human subjects: a randomized trial
- Author
-
Jakob Hendel, Maria Juul Nielsen, Alfredo Rago, Jesper A.B. Strickertsson, Albin Sandelin, Lars H. Engelholm, Kristoffer Vitting-Seerup, Christa Broholm, Christina V Müller, Adam Baker, Cathrine Melsæther, Kim B. Jensen, Stine Sloth, Yun Chen, and Jette Bornholdt
- Subjects
0301 basic medicine ,Microbiology (medical) ,Adult ,DNA, Bacterial ,Male ,human transcriptomics ,Gene Expression ,Lymphocyte Activation ,Microbiology ,law.invention ,03 medical and health sciences ,Probiotic ,Young Adult ,fluids and secretions ,0302 clinical medicine ,Human gut ,Sex Factors ,Lactobacillus rhamnosus ,Randomized controlled trial ,law ,lactobacillus rhamnosus gg ,medicine ,Humans ,lcsh:RC799-869 ,B cell ,B-cell activation ,B-Lymphocytes ,Cross-Over Studies ,biology ,immediate in vivo effect ,Lacticaseibacillus rhamnosus ,Gastroenterology ,food and beverages ,b cell activation ,biology.organism_classification ,Healthy Volunteers ,Gastrointestinal Microbiome ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Jejunum ,Gene Expression Regulation ,probiotics ,030211 gastroenterology & hepatology ,Female ,lcsh:Diseases of the digestive system. Gastroenterology ,Research Article ,Research Paper - Abstract
The specific effects of administering live probiotics in the human gut are not well characterized. To this end, we investigated the immediate effect of Lactobacillus rhamnosus GG (LGG) in the jejunum of 27 healthy volunteers 2 h after ingestion using a combination of global RNA sequencing of human biopsies and bacterial DNA sequencing in a multi-visit, randomized, cross-over design (ClinicalTrials.gov number NCT03140878). While LGG was detectable in jejunum after 2 h in treated subjects, the gene expression response vs. placebo was subtle if assessed across all subjects. However, clustering analysis revealed that one-third of subjects exhibited a strong and consistent LGG response involving hundreds of genes, where genes related to B cell activation were upregulated, consistent with prior results in mice. Immunohistochemistry and single cell-based deconvolution analyses showed that this B cell signature likely is due to activation and proliferation of existing B cells rather than B cell immigration to the tissue. Our results indicate that the LGG strain has an immediate effect in the human gut in a subpopulation of individuals. In extension, our data strongly suggest that studies on in vivo probiotic effects in humans require large cohorts and must take individual variation into account.
- Published
- 2020
- Full Text
- View/download PDF
27. Immune regulation by fibroblasts in tissue injury depends on uPARAP-mediated uptake of collectins
- Author
-
Niels Behrendt, Lars H. Engelholm, Katharina Wassilew, Kirstine S Nørregaard, Megan M. Sibree, Henrik J. Jürgensen, Daniel H. Madsen, Thomas H. Bugge, Dorrit Krustrup, Eric Santoni-Rugiu, and Peter Garred
- Subjects
Pulmonary Fibrosis ,Endocytic cycle ,Gene Expression ,Collectin ,Mice, Transgenic ,Receptors, Cell Surface ,Biology ,Lung injury ,Mannose-Binding Lectin ,Article ,Collagen receptor ,Bleomycin ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Dermis ,medicine ,Animals ,Humans ,Lectins, C-Type ,Secretion ,Receptor ,Lung ,Research Articles ,030304 developmental biology ,0303 health sciences ,Membrane Glycoproteins ,Interleukin-6 ,Surfactant protein D ,Lung Injury ,Cell Biology ,Fibroblasts ,Pulmonary Surfactant-Associated Protein D ,Survival Analysis ,Endocytosis ,Immunity, Innate ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,Mannose-Binding Lectins ,medicine.anatomical_structure ,Proteolysis ,Lysosomes ,Bronchoalveolar Lavage Fluid ,Mannose Receptor ,030217 neurology & neurosurgery - Abstract
Tissue injury can lead to fibrotic disease and chronic inflammation, for example, in the lungs of patients with idiopathic pulmonary fibrosis. Jürgensen et al. report that fibroblasts regulate immune responses during tissue injury by clearing collectins with pro- or antiinflammatory activity via uPARAP, an endocytic collectin receptor identified in this work., Collectins such as mannose-binding lectin (MBL) and surfactant protein D (SP-D) become temporarily deposited in extravascular compartments after tissue injury and perform immune-stimulatory or inflammation-limiting functions. However, their turnover mechanisms, necessary to prevent excessive tissue damage, are virtually unknown. In this study, we show that fibroblasts in injured tissues undertake the clearance of collectins by using the endocytic collagen receptor uPARAP. In cellular assays, several types of collectins were endocytosed in a highly specific uPARAP-dependent process, not shared by the closely related receptor MR/CD206. When introduced into dermis or bleomycin-injured lungs of mice, collectins MBL and SP-D were endocytosed and routed for lysosomal degradation by uPARAP-positive fibroblasts. Fibroblast-specific expression of uPARAP governed endogenous SP-D levels and overall survival after lung injury. In lung tissue from idiopathic pulmonary fibrosis patients, a strong up-regulation of uPARAP was observed in fibroblasts adjacent to regions with SP-D secretion. This study demonstrates a novel immune-regulatory function of fibroblasts and identifies uPARAP as an endocytic receptor in immunity.
- Published
- 2018
- Full Text
- View/download PDF
28. Phagocytosis of collagen fibrils by fibroblasts in vivo is independent of the uPARAP/Endo180 receptor
- Author
-
Lars H. Engelholm, Sara Sprangers, Yixuan Cao, Niels Behrendt, Vincent Everts, Orale Celbiologie (ORM, ACTA), ACTA, Oral Cell Biology, and Academic Centre for Dentistry Amsterdam
- Subjects
0301 basic medicine ,Periodontal Ligament ,Fibrillar Collagens ,Phagocytosis ,Receptors, Cell Surface ,Biochemistry ,Collagen receptor ,Extracellular matrix ,Mice ,03 medical and health sciences ,Microscopy, Electron, Transmission ,Periosteum ,Extracellular ,medicine ,Animals ,Fibroblast ,Molecular Biology ,Membrane Glycoproteins ,Tibia ,Chemistry ,Skull ,Fibrillogenesis ,Cell Biology ,Fibroblasts ,Molar ,Extracellular Matrix ,Cell biology ,Incisor ,Collagen, type I, alpha 1 ,030104 developmental biology ,medicine.anatomical_structure ,Intracellular - Abstract
As a crucial step in ECM remodeling, collagen degradation occurs through different processes, including both extracellular and intracellular degradation. The extracellular pathways of collagen degradation require secretion of collagenolytic proteases, whereas intracellular collagen degradation occurs in the lysosomal compartment after uptake, involving either pre-cleaved or intact fibrillar collagen. The endocytic collagen receptor uPARAP/Endo180 plays an important role in internalization of large collagen degradation products, whereas its role in the phagocytosis of fibrillar collagen has been debated. In fact, the role of this receptor in regular collagen phagocytosis in vivo has not been established. In this study, we have studied the role of uPARAP in the phagocytosis of collagen fibrils in vivo by analyzing different connective tissues of mice lacking uPARAP. Using transmission electron microscopy (TEM), we found that fibroblasts in the periosteum of tibia and calvaria, as well as in the periodontal ligament of molar and incisor, phagocytosed collagen fibrils independently of uPARAP. Quantification of phagocytosed collagen in the periodontal ligament of uPARAP-deficient mice and controls revealed no difference in the amount of fibrillar collagen taken up by uPARAP-deficient mice. The findings show that under in vivo conditions uPARAP does not play a role in the phagocytic uptake of collagen fibrils by fibroblasts. Consequently, the cellular uptake of collagen fibrils and collagen cleavage products probably occurs through fundamentally different pathways. J. Cell. Biochem. 118: 1590-1595, 2017. © 2016 Wiley Periodicals, Inc.
- Published
- 2017
- Full Text
- View/download PDF
29. Cellular uptake of collagens and implications for immune cell regulation in disease
- Author
-
Kirstine S Nørregaard, Niels Behrendt, Thomas H. Bugge, Henrik J. Jürgensen, Daniel H. Madsen, Lars H. Engelholm, and Sander van Putten
- Subjects
Pharmacology ,0303 health sciences ,Innate immune system ,Chemistry ,030302 biochemistry & molecular biology ,Endocytic cycle ,Cell Biology ,Fibrosis ,Endocytosis ,Collagen receptor ,Cell biology ,Extracellular Matrix ,Extracellular matrix ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Immune system ,Neoplasms ,Extracellular ,Molecular Medicine ,Animals ,Humans ,Collagen ,Cell adhesion ,Molecular Biology ,Mannose receptor - Abstract
As the dominant constituent of the extracellular matrix (ECM), collagens of different types are critical for the structural properties of tissues and make up scaffolds for cellular adhesion and migration. Importantly, collagens also directly modulate the phenotypic state of cells by transmitting signals that influence proliferation, differentiation, polarization, survival, and more, to cells of mesenchymal, epithelial, or endothelial origin. Recently, the potential of collagens to provide immune regulatory signals has also been demonstrated, and it is believed that pathological changes in the ECM shape immune cell phenotype. Collagens are themselves heavily regulated by a multitude of structural modulations or by catabolic pathways. One of these pathways involves a cellular uptake of collagens or soluble collagen-like defense collagens of the innate immune system mediated by endocytic collagen receptors. This cellular uptake is followed by the degradation of collagens in lysosomes. The potential of this pathway to regulate collagens in pathological conditions is evident from the increased extracellular accumulation of both collagens and collagen-like defense collagens following endocytic collagen receptor ablation. Here, we review how endocytic collagen receptors regulate collagen turnover during physiological conditions and in pathological conditions, such as fibrosis and cancer. Furthermore, we highlight the potential of collagens to regulate immune cells and discuss how endocytic collagen receptors can directly regulate immune cell activity in pathological conditions or do it indirectly by altering the extracellular milieu. Finally, we discuss the potential collagen receptors utilized by immune cells to directly detect ECM-related changes in the tissues which they encounter.
- Published
- 2019
30. Collagen density regulates the activity of tumor-infiltrating T cells
- Author
-
Adrija Kalvisa, Anne Mette Hvid Larsen, Lars H. Engelholm, Dorota Ewa Kuczek, Elfriede Noessner, Ana Micaela Carnaz Simoes, Majken S. Siersbæk, Marco Carretta, Marco Donia, Per thor Straten, Daniel H. Madsen, Anne Roslind, Marie-Louise Thorseth, Inge Marie Svane, and Lars Grøntved
- Subjects
0301 basic medicine ,3D culture ,Cancer Research ,Regulatory T cell ,T cell ,Immunology ,Immune modulation ,Lymphocyte Activation ,lcsh:RC254-282 ,Extracellular matrix ,Immunomodulation ,03 medical and health sciences ,0302 clinical medicine ,Lymphocytes, Tumor-Infiltrating ,Cell Line, Tumor ,Neoplasms ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Humans ,Autocrine signalling ,Cells, Cultured ,030304 developmental biology ,Cell Proliferation ,Pharmacology ,0303 health sciences ,Tumor microenvironment ,T cell activity ,Chemistry ,Gene Expression Profiling ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3d Culture ,Extracellular Matrix ,Immune Modulation ,T Cell Activity ,Tumor Microenvironment ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Molecular Medicine ,Collagen ,CD8 ,Research Article - Abstract
BackgroundTumor progression is accompanied by dramatic remodeling of the surrounding extracellular matrix leading to the formation of a tumor-specific ECM, which is often more collagen-rich and of increased stiffness. The altered ECM of the tumor supports cancer growth and metastasis, but it is unknown if this effect involves modulation of T cell activity. To investigate if a high-density tumor-specific ECM could influence the ability of T cells to kill cancer cells, we here studied how T cells respond to 3D culture in different collagen densities.MethodsT cells cultured in 3D conditions surrounded by a high or low collagen density were imaged using confocal fluorescent microscopy. The effects of the different collagen densities on T cell proliferation, survival, and differentiation were examined using flow cytometry. Cancer cell proliferation in similar 3D conditions was also measured. Triple-negative breast cancer specimens were analyzed for the number of infiltrating CD8+ T cells and for the collagen density. Whole-transcriptome analyses were applied to investigate in detail the effects of collagen density on T cells. Computational analyses were used to identify transcription factors involved in the collagen density-induced gene regulation. Observed changes were confirmed by qRT-PCR analysis.ResultsT cell proliferation was significantly reduced in a high-density matrix compared to a low-density matrix and prolonged culture in a high-density matrix led to a higher ratio of CD4+ to CD8+ T cells. The proliferation of cancer cells was unaffected by the surrounding collagen-density. Consistently, we observed a reduction in the number of infiltrating CD8+ T-cells in mammary tumors with high collagen-density indicating that collagen-density has a role in regulating T cell abundance in human breast cancer.Whole-transcriptome analysis of 3D-cultured T cells revealed that a high-density matrix induces downregulation of cytotoxic activity markers and upregulation of regulatory T cell markers. These transcriptional changes were predicted to involve autocrine TGF-B signaling and they were accompanied by an impaired ability of tumor-infiltrating T cells to kill autologous cancer cells.ConclusionsOur study identifies a new immune modulatory mechanism, which could be essential for suppression of T cell activity in the tumor microenvironment.
- Published
- 2019
- Full Text
- View/download PDF
31. The focal adhesion-associated proteins DOCK5 and GIT2 comprise a rheostat in control of epithelial invasion
- Author
-
Scott R. Frank, Clemens P. Köllmann, Steen H. Hansen, Jay R. Thiagarajah, J F van Lidth de Jeude, Morten Frödin, and Lars H. Engelholm
- Subjects
0301 basic medicine ,Cancer Research ,GTPase-activating protein ,Gene Expression ,GTPase ,CDC42 ,Models, Biological ,Focal adhesion ,03 medical and health sciences ,Adapter molecule crk ,Mice ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,Genetics ,Cell Adhesion ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Neoplasm Metastasis ,RNA, Small Interfering ,Cell adhesion ,Molecular Biology ,biology ,Dock5 ,GTPase-Activating Proteins ,Cell biology ,Disease Models, Animal ,Protein Transport ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Heterografts ,Original Article ,Female ,Guanine nucleotide exchange factor ,Acute-Phase Proteins ,HeLa Cells ,Protein Binding - Abstract
DOCK proteins are guanine nucleotide exchange factors for Rac and Cdc42 GTPases. DOCK1 is the founding member of the family and acts downstream of integrins via the canonical Crk-p130Cas complex to activate Rac GTPases in numerous contexts. In contrast, DOCK5, which possesses the greatest similarity to DOCK1, remains sparingly studied. Here we establish that DOCK5 has a non-redundant role in regulating motile and invasive capacities of epithelial cells. DOCK1 is constitutively associated with sites of integrin attachment termed focal adhesions (FAs). In contrast, we demonstrate that DOCK5 recruitment to FAs in Hela cells is restricted by GIT2, an established regulator of FA signaling. We determine that GIT2 is targeted to FAs in response to Rho-ROCK signaling and actomyosin contractility. Accordingly, inhibition of ROCK activity or MLC function promotes enrichment of DOCK5 in membrane protrusions and nascent cell-substratum adhesions. We further demonstrate that GIT2 inhibits the interaction of DOCK5 with Crk. Moreover, we show that depletion of GIT2 promotes DOCK5-dependent activation of the Crk-p130Cas signaling cascade to promote Rac1-mediated lamellipodial protrusion and FA turnover. The antagonism between GIT2 and DOCK5 extends to non-transformed MCF10A mammary epithelial cells, with DOCK5 'dialing-up' and GIT2 'dialing-down' invasiveness. Finally, we determine that DOCK5 inhibition attenuates invasion and metastasis of MDA-MB-231 cells and prolongs life span of mice injected with these cells. Collectively, our work identifies DOCK5 as a key regulator of epithelial invasion and metastasis, and demonstrates that suppression of DOCK5 by GIT2 represents a previously unappreciated mechanism for coordination of Rho and Rac GTPases.Oncogene advance online publication, 26 September 2016; doi:10.1038/onc.2016.345.
- Published
- 2016
- Full Text
- View/download PDF
32. Early reversal cells in adult human bone remodeling: osteoblastic nature, catabolic functions and interactions with osteoclasts
- Author
-
Maja Hinge, Thomas Levin Andersen, Lars H. Engelholm, Lars Rolighed, Pia Rosgaard Jensen, Niels Marcussen, Jean-Marie Delaissé, Mohamed Essameldin Abdelgawad, and Ragad Walid Alnaimi
- Subjects
Adult ,Male ,musculoskeletal diseases ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Histology ,Interaction ,Osteoclasts ,030209 endocrinology & metabolism ,Bone resorption ,Bone remodeling ,03 medical and health sciences ,0302 clinical medicine ,Microscopy, Electron, Transmission ,Reversal phase ,Osteoclast ,Collagenolysis ,Bone cell ,medicine ,Humans ,Molecular Biology ,Aged ,Osteoblasts ,Osteoid ,Chemistry ,Osteoblast ,Reversal cells ,Catabolism ,Cell Biology ,Middle Aged ,Hyperparathyroidism, Primary ,Resorption ,Cell biology ,Medical Laboratory Technology ,030104 developmental biology ,medicine.anatomical_structure ,Female ,Bone Remodeling ,Cancellous bone - Abstract
The mechanism coupling bone resorption and formation is a burning question that remains incompletely answered through the current investigations on osteoclasts and osteoblasts. An attractive hypothesis is that the reversal cells are likely mediators of this coupling. Their nature is a big matter of debate. The present study performed on human cancellous bone is the first one combining in situ hybridization and immunohistochemistry to demonstrate their osteoblastic nature. It shows that the Runx2 and CD56 immunoreactive reversal cells appear to take up TRAcP released by neighboring osteoclasts. Earlier preclinical studies indicate that reversal cells degrade the organic matrix left behind by the osteoclasts and that this degradation is crucial for the initiation of the subsequent bone formation. To our knowledge, this study is the first addressing these catabolic activities in adult human bone through electron microscopy and analysis of molecular markers. Periosteoclastic reversal cells show direct contacts with the osteoclasts and with the demineralized resorption debris. These early reversal cells show (1) ¾-collagen fragments typically generated by extracellular collagenases of the MMP family, (2) MMP-13 (collagenase-3) and (3) the endocytic collagen receptor uPARAP/Endo180. The prevalence of these markers was lower in the later reversal cells, which are located near the osteoid surfaces and morphologically resemble mature bone-forming osteoblasts. In conclusion, this study demonstrates that reversal cells colonizing bone surfaces right after resorption are osteoblast-lineage cells, and extends to adult human bone remodeling their role in rendering eroded surfaces osteogenic.
- Published
- 2016
- Full Text
- View/download PDF
33. Targeting a novel bone degradation pathway in primary bone cancer by inactivation of the collagen receptor uPARAP/Endo180
- Author
-
Signe Ingvarsen, Clement S. Trovik, Andreas Hald, Kirstine S Nørregaard, Ole Didrik Laerum, Niels Behrendt, Johan Eide, Morten Persson, Thomas H. Bugge, Henrik J. Jürgensen, Christoffer Nielsen, Daniel H. Madsen, Kristina Johansson, Maria C. Melander, Andreas Kjaer, and Lars H. Engelholm
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Osteolysis ,Bone cancer ,Mesenchymal stem cell ,Cancer ,Biology ,medicine.disease ,3. Good health ,Pathology and Forensic Medicine ,Collagen receptor ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Primary bone ,030220 oncology & carcinogenesis ,medicine ,Osteosarcoma ,Sarcoma - Abstract
In osteosarcoma, a primary mesenchymal bone cancer occurring predominantly in younger patients, invasive tumour growth leads to extensive bone destruction. This process is insufficiently understood, cannot be efficiently counteracted and calls for novel means of treatment. The endocytic collagen receptor, uPARAP/Endo180, is expressed on various mesenchymal cell types and is involved in bone matrix turnover during normal bone growth. Human osteosarcoma specimens showed strong expression of this receptor on tumour cells, along with the collagenolytic metalloprotease, MT1-MMP. In advanced tumours with ongoing bone degeneration, sarcoma cells positive for these proteins formed a contiguous layer aligned with the degradation zones. Remarkably, osteoclasts were scarce or absent from these regions and quantitative analysis revealed that this scarcity marked a strong contrast between osteosarcoma and bone metastases of carcinoma origin. This opened the possibility that sarcoma cells might directly mediate bone degeneration. To examine this question, we utilized a syngeneic, osteolytic bone tumour model with transplanted NCTC-2472 sarcoma cells in mice. When analysed in vitro, these cells were capable of degrading the protein component of surface-labelled bone slices in a process dependent on MMP activity and uPARAP/Endo180. Systemic treatment of the sarcoma-inoculated mice with a mouse monoclonal antibody that blocks murine uPARAP/Endo180 led to a strong reduction of bone destruction. Our findings identify sarcoma cell-resident uPARAP/Endo180 as a central player in the bone degeneration of advanced tumours, possibly following an osteoclast-mediated attack on bone in the early tumour stage. This points to uPARAP/Endo180 as a promising therapeutic target in osteosarcoma, with particular prospects for improved neoadjuvant therapy.
- Published
- 2015
- Full Text
- View/download PDF
34. Microvessel density and endothelial cell proliferation levels in colorectal liver metastases from patients given neo-adjuvant cytotoxic chemotherapy and bevacizumab
- Author
-
Hans Christian Rolff, Lars H. Engelholm, Gro Linno Willemoe, Kell Østerlind, Ib Jarle Christensen, Ole Didrik Laerum, Rikke Løvendahl Eefsen, Martin Illemann, Ben Vainer, Gunilla Høyer-Hansen, and Gert Van den Eynden
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,Bevacizumab ,Colorectal cancer ,business.industry ,Angiogenesis ,medicine.medical_treatment ,Combination chemotherapy ,medicine.disease ,Metastasis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Adenocarcinoma ,business ,Microvessel ,medicine.drug - Abstract
The treatment of patients with colorectal liver metastasis has improved significantly and first line therapy is often combined chemotherapy and bevacizumab, although it is unknown who responds to this regimen. Colorectal liver metastases grow in different histological growth patterns showing differences in angiogenesis. To identify possible response markers, histological markers of angiogenesis were assessed. Patients who underwent resection of colorectal liver metastasis at Rigshospitalet, Copenhagen, Denmark from 2007 to 2011 were included (n = 254) including untreated and patients treated with chemotherapy or chemotherapy plus bevacizumab. The resected liver metastases were characterised with respect to growth pattern, endothelial and tumour cell proliferation as well as microvessel density and tumour regression. Tumour regression grade of liver metastases differed significantly between untreated/chemotherapy treated patients in comparison to chemotherapy plus bevacizumab treated patients (both p
- Published
- 2015
- Full Text
- View/download PDF
35. CRISPR/Cas9 Engineering of Adult Mouse Liver Demonstrates That the Dnajb1-Prkaca Gene Fusion is Sufficient to Induce Tumors Resembling Fibrolamellar Hepatocellular Carcinoma
- Author
-
Steen H. Hansen, Francesco Niola, Morten Frödin, Lars H. Engelholm, Jens Vilstrup Johansen, Anjum Riaz, Eric Santoni-Rugiu, Denise Serra, and Frederik Dagnæs-Hansen
- Subjects
Genomic Engineering ,Liver Cancer ,0301 basic medicine ,Carcinoma, Hepatocellular ,Time Factors ,Liver tumor ,CRISPR-Associated Proteins ,Biology ,Article ,Fusion gene ,Mice ,03 medical and health sciences ,Exon ,Liver disease ,0302 clinical medicine ,Biomarkers, Tumor ,medicine ,Journal Article ,Protein Kinase A ,Animals ,PKA ,Clustered Regularly Interspaced Short Palindromic Repeats ,Genetic Predisposition to Disease ,Gene ,Gene Editing ,Cyclic AMP-Dependent Protein Kinase Catalytic Subunits ,Mouse Model ,Hepatology ,Liver Neoplasms ,Gastroenterology ,HSP40 Heat-Shock Proteins ,medicine.disease ,Molecular biology ,PRKACA ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,Phenotype ,030104 developmental biology ,Fibrolamellar hepatocellular carcinoma ,030220 oncology & carcinogenesis ,Disease Progression ,Cancer research ,Female ,CRISPR-Cas Systems ,Gene Fusion ,Liver cancer - Abstract
Background & Aims Fibrolamellar hepatocellular carcinoma (FL-HCC) is a primary liver cancer that predominantly affects children and young adults with no underlying liver disease. A somatic, 400 Kb deletion on chromosome 19 that fuses part of the DnaJ heat shock protein family (Hsp40) member B1 gene ( DNAJB1 ) to the protein kinase cAMP-activated catalytic subunit alpha gene ( PRKACA ) has been repeatedly identified in patients with FL-HCC. However, the DNAJB1 – PRKACA gene fusion has not been shown to induce liver tumorigenesis. We used the CRISPR/Cas9 technique to delete in mice the syntenic region on chromosome 8 to create a Dnajb1–Prkaca fusion and monitored the mice for liver tumor development. Methods We delivered CRISPR/Cas9 vectors designed to juxtapose exon 1 of Dnajb1 with exon 2 of Prkaca to create the Dnajb1–Prkaca gene fusion associated with FL-HCC, or control Cas9 vector, via hydrodynamic tail vein injection to livers of 8-week-old female FVB/N mice. These mice did not have any other engineered genetic alterations and were not exposed to liver toxins or carcinogens. Liver tissues were collected 14 months after delivery; genomic DNA was analyzed by PCR to detect the Dnajb1–Prkaca fusion, and tissues were characterized by histology, immunohistochemistry, RNA sequencing, and whole-exome sequencing. Results Livers from 12 of the 15 mice given the vectors to induce the Dnajb1–Prkaca gene fusion, but none of the 11 mice given the control vector, developed neoplasms. The tumors contained the Dnajb1–Prkaca gene fusion and had histologic and cytologic features of human FL-HCCs: large polygonal cells with granular, eosinophilic, and mitochondria-rich cytoplasm, prominent nucleoli, and markers of hepatocytes and cholangiocytes. In comparing expression levels of genes between the mouse tumor and non-tumor liver cells, we identified changes similar to those detected in human FL-HCC, which included genes that affect cell cycle and mitosis regulation. Genomic analysis of mouse neoplasms induced by the Dnajb1–Prkaca fusion revealed a lack of mutations in genes commonly associated with liver cancers, as observed in human FL-HCC. Conclusions Using CRISPR/Cas9 technology, we found generation of the Dnajb1–Prkaca fusion gene in wild-type mice to be sufficient to initiate formation of tumors that have many features of human FL-HCC. Strategies to block DNAJB1–PRKACA might be developed as therapeutics for this form of liver cancer.
- Published
- 2017
- Full Text
- View/download PDF
36. The collagen receptor uPARAP/Endo180 as a novel target for antibody-drug conjugate mediated treatment of mesenchymal and leukemic cancers
- Author
-
Petra Hamerlik, Kirstine S Nørregaard, Kamilla E. Jensen, Signe Ingvarsen, Christoffer Nielsen, Sander van Putten, Lars H. Engelholm, Kristine Rothaus Christensen, Henrik Gårdsvoll, Ida K. Lund, Maria C. Melander, Kristian Reckzeh, Henrik J. Jürgensen, and Niels Behrendt
- Subjects
0301 basic medicine ,Antibody-drug conjugate ,Immunoconjugates ,sarcoma ,medicine.drug_class ,Cell Survival ,Gene Expression ,Receptors, Cell Surface ,Monoclonal antibody ,Collagen receptor ,antibody-drug conjugate ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Antineoplastic Agents, Immunological ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,uPARAP ,Leukemia ,Membrane Glycoproteins ,biology ,business.industry ,Cell Cycle ,glioblastoma ,Cancer ,Cell cycle ,medicine.disease ,Xenograft Model Antitumor Assays ,Endocytosis ,Tumor Burden ,Disease Models, Animal ,030104 developmental biology ,Mannose-Binding Lectins ,Oncology ,Monomethyl auristatin E ,chemistry ,Receptors, Mitogen ,Immunology ,biology.protein ,Cancer research ,Antibody ,business ,Research Paper - Abstract
A key task in developing the field of personalized cancer therapy is the identification of novel molecular targets that enable treatment of cancers not susceptible to other means of specific therapy. The collagen receptor uPARAP/Endo180 is overexpressed by malignant cells in several non-epithelial cancers, notably including sarcomas, glioblastomas and subsets of acute myeloid leukemia. In contrast, in healthy adult individuals, expression is restricted to minor subsets of mesenchymal cells. Functionally, uPARAP/Endo180 is a rapidly recycling endocytic receptor that delivers its cargo directly into the endosomal-lysosomal system, thus opening a potential route of entry into receptor-positive cells. This combination of specific expression and endocytic function appears well suited for targeting of uPARAP/Endo180-positive cancers by antibody-drug conjugate (ADC) mediated drug delivery. Therefore, we utilized a specific monoclonal antibody against uPARAP/Endo180, raised through immunization of a uPARAP/Endo180 knock-out mouse, which reacts with both the human and the murine receptor, to construct a uPARAP-directed ADC. This antibody was coupled to the highly toxic dolastatin derivative, monomethyl auristatin E, via a cathepsin-labile valine-citrulline linker. With this ADC, we show strong and receptor-dependent cytotoxicity in vitro in uPARAP/Endo180-positive cancer cell lines of sarcoma, glioblastoma and leukemic origin. Furthermore, we demonstrate the potency of the ADC in vivo in a xenograft mouse model with human uPARAP/Endo180-positive leukemic cells, obtaining a complete cure of all tested mice following intravenous ADC treatment with no sign of adverse effects. Our study identifies uPARAP/Endo180 as a promising target for novel therapy against several highly malignant cancer types.
- Published
- 2017
- Full Text
- View/download PDF
37. Monitoring mammary tumor progression and effect of tamoxifen treatment in MMTV-PymT using MRI and magnetic resonance spectroscopy with hyperpolarized [1-13 C]pyruvate
- Author
-
Olaf B. Paulson, Per Åkeson, Peter Magnusson, Susanne Holck, Jan Henrik Ardenkjær-Larsen, Lise Vejby Søgaard, Lars H. Engelholm, Sadia Asghar Butt, Mette Hauge Lauritzen, and Osman Mirza
- Subjects
Pathology ,medicine.medical_specialty ,Mammary tumor ,Metabolism ,medicine.disease ,In vitro ,chemistry.chemical_compound ,Breast cancer ,chemistry ,In vivo ,Lactate dehydrogenase ,medicine ,Radiology, Nuclear Medicine and imaging ,Ex vivo ,Tamoxifen ,medicine.drug - Abstract
Purpose To use dynamic magnetic resonance spectroscopy (MRS) of hyperpolarized 13C-pyruvate to follow the progress over time in vivo of breast cancer metabolism in the MMTV-PymT model, and to follow the response to the anti-estrogen drug tamoxifen. Methods Tumor growth was monitored by anatomical MRI by measuring tumor volumes. Dynamic MRS of hyperpolarized 13C was used to measure an “apparent” pyruvate-to-lactate rate constant (kp) of lactate dehydrogenase (LDH) in vivo. Further, ex vivo pathology and in vitro LDH initial reaction velocity were evaluated. Results Tamoxifen significantly halted the tumor growth measured as tumor volume by MRI. In the untreated animals, kp correlated with tumor growth. The kP was somewhat but not significantly lower in the treated group. Studies in vitro confirmed the effects of tamoxifen on tumor growth, and here the LDH reaction velocity was reduced significantly in the treated group. Conclusion These hyperpolarized 13C MRS findings indicate that tumor metabolic changes affects kP. The measured kp did not relate to treatment response to the same extent as did tumor growth, histological evaluation, and in vitro determination of LDH activity. Magn Reson Med 73:51–58, 2015. © 2014 Wiley Periodicals, Inc.
- Published
- 2014
- Full Text
- View/download PDF
38. CCL2/MCP-1 signaling drives extracellular matrix turnover by diverse macrophage subsets
- Author
-
Oliver Krigslund, Thomas H. Bugge, Lakmali Munasinghage Silva, Daniel H. Madsen, Niels Behrendt, Lars H. Engelholm, Sander van Putten, and Henrik J. Jürgensen
- Subjects
Extracellular matrix endocytosis ,Histology ,medicine.medical_treatment ,Biophysics ,Plg, plasminogen ,CEMS, collagen-endocytosing macrophages ,CCL2 ,Endocytosis ,Biochemistry ,Article ,M-CSF, Macrophage-Colony Stimulating Factor ,IL4Ra, IL4 Receptor a ,Flow cytometry ,Proinflammatory cytokine ,AF, Alexa Fluor ,Extracellular matrix ,RFP, red fluorescent protein ,CCL2/MCP-1, chemokine (C-C motif) ligand 2/monocyte chemoattractant protein 1 ,Dermis ,GM-CSF, Granulocyte Macrophage-Colony Stimulating Factor ,Genetics ,medicine ,Macrophage ,IL, Interleukin ,FMO, fluorescence minus one ,Fibrin degradation ,lcsh:QH301-705.5 ,Molecular Biology ,Mannose receptor/CD206 ,Interleukin-13 ,medicine.diagnostic_test ,Chemistry ,MR, mannose receptor/CD206 ,Cell Biology ,Collagen degradation ,ECM, extracellular matrix ,uPARAP, urokinase plasminogen activator receptor associated protein/Endo180 ,Cell biology ,Cytokine ,medicine.anatomical_structure ,lcsh:Biology (General) ,GM-CSFR, GM-CSF Receptor ,CCR2, C-C chemokine receptor type 2 ,FEMS, fibrin-endocytosing macrophages ,uPARAP/Endo180 - Abstract
Macrophage plasticity, cellular origin, and phenotypic heterogeneity are perpetual challenges for studies addressing the biology of this pivotal immune cell in development, homeostasis, and tissue remodeling/repair. Consequently, a myriad of macrophage subtypes has been described in these contexts. To facilitate the identification of functional macrophage subtypes in vivo, here we used a flow cytometry-based assay that allows for detailed phenotyping of macrophages engaged in extracellular matrix (ECM) degradation. Of the five macrophage subtypes identified in the remodeling dermis by using this assay, collagen degradation was primarily executed by Ly6C−CCR2+ and Ly6C−CCR2low macrophages via mannose receptor-dependent collagen endocytosis, while Ly6C+CCR2+ macrophages were the dominant fibrin-endocytosing cells. Unexpectedly, the CCL2/MCP1-CCR2 signaling axis was critical for both collagen and fibrin degradation, while collagen degradation was independent of IL-4Ra signaling. Furthermore, the cytokine GM-CSF selectively enhanced collagen degradation by Ly6C+CCR2+ macrophages. This study reveals distinct subsets of macrophages engaged in ECM turnover and identifies novel wound healing-associated functions for CCL2 and GM-CSF inflammatory cytokines., Highlights • Phenotypically diverse subsets of dermal macrophages undertake the degradation of extracellular matrix • C-C motif chemokine Ligand 2 (CCL2) signaling is critical for macrophage-mediated endocytosis of collagen and fibrin. • Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) and Interleukin (IL)-13 stimulate collagen endocytosis. • The wound healing-associated IL4-IL4 Receptor a (IL4Ra) signaling is dispensable for collagen endocytosis by macrophages. • The mannose receptor is the principal endocytic collagen receptor utilized by resident dermal macrophages.
- Published
- 2019
- Full Text
- View/download PDF
39. The number of regulatory T cells in transbronchial lung allograft biopsies is related to FoxP3 mRNA levels in bronchoalveolar lavage fluid and to the degree of acute cellular rejection
- Author
-
Dorrit Krustrup, Caroline B. Madsen, Lars H. Engelholm, Martin Iversen, Lars P. Ryder, and Claus B. Andersen
- Subjects
Adult ,Graft Rejection ,Male ,Pathology ,medicine.medical_specialty ,Regulatory T cell ,medicine.medical_treatment ,Immunology ,chemical and pharmacologic phenomena ,Bronchoalveolar Lavage ,T-Lymphocytes, Regulatory ,Immune system ,Antigen ,Humans ,Immunology and Allergy ,Medicine ,Lung transplantation ,RNA, Messenger ,Lung ,Retrospective Studies ,Immunity, Cellular ,Transplantation ,medicine.diagnostic_test ,business.industry ,FOXP3 ,Forkhead Transcription Factors ,hemic and immune systems ,Middle Aged ,respiratory system ,Allografts ,respiratory tract diseases ,Bronchoalveolar lavage ,medicine.anatomical_structure ,Immunohistochemistry ,Female ,business ,Lung Transplantation - Abstract
Background The transcription factor Forkhead Box P3 (FoxP3) is a marker of regulatory T cells (Tregs) — a subset of T cells known to suppress a wide range of immune responses. These cells are considered to be pivotal for the induction of tolerance to donor antigens in human allografts. We aimed to correlate the number of lymphocytes expressing FoxP3 in transbronchial biopsies from lung allografts with the FoxP3 expression in bronchoalveolar lavage fluid (BALF). In addition, we aimed to correlate the number of FoxP3 + cells in transbronchial biopsies with the degree of acute cellular rejection in lung allografts. Materials and methods The expression of FoxP3 was evaluated using immunohistochemical staining in 40 lung allograft biopsies obtained from 23 patients. The number of Tregs was related to the FoxP3 mRNA levels as determined using qRT-PCR in corresponding BALF samples from the same patients. Furthermore, the number of Tregs was related to the degree of acute allograft rejection (according to ISHLT criteria, A0–A4). Results Regression analysis showed a significant concordance between the number of Tregs in lung tissue and the level of FoxP3 mRNA relative to glyceraldehyde 3-phosphate dehydrogenase (GAPDH) mRNA levels in BALF (n = 40, p = 0.0001). In addition, we found a significant increase in the number of Tregs during acute allograft rejections of grades A2 and higher (median: 32.6 Tregs/mm 2 ) when compared to those of grades A1 and A0 (median: 4.9 Tregs/mm 2 ) (p = 0.0002). Discussion and conclusion The association between the distribution of Tregs in transbronchial biopsies and the level of FoxP3 mRNA in BALF indicates that assessment of FoxP3 mRNA in BALF is a reliable non-invasive method for evaluating the number of Tregs in lung tissue. Furthermore, the association between the number of Tregs in lung tissue and the degree of acute cellular rejection shows that Tregs are recruited to the site of inflammation and may be involved in the regulation of acute rejection. Thus, Tregs may play a role in the cellular processes that affect lung allograft outcome.
- Published
- 2013
- Full Text
- View/download PDF
40. Targeting a Single Function of the Multifunctional Matrix Metalloprotease MT1-MMP
- Author
-
Signe Ingvarsen, Daniel H. Madsen, Niels Behrendt, Ludovic Maertens, Henrik Gårdsvoll, Astrid Porse, Maria C. Melander, Charlotte Erpicum, Lars H. Engelholm, Agnès Noël, Henrik J. Jürgensen, Gunilla Høyer-Hansen, and Kenn Holmbeck
- Subjects
0303 health sciences ,Angiogenesis ,Chemistry ,Cell Biology ,Multifunctional Enzymes ,Matrix metalloproteinase ,Matrix (biology) ,Biochemistry ,Molecular biology ,3. Good health ,Lymphangiogenesis ,Cell biology ,Extracellular matrix ,03 medical and health sciences ,Enzyme activator ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Gelatinase ,Molecular Biology ,030304 developmental biology - Abstract
The group of matrix metalloproteases (MMPs) is responsible for multiple processes of extracellular matrix remodeling in the healthy body but also for matrix and tissue destruction during cancer invasion and metastasis. The understanding of the contributions from each individual MMP, both in healthy and pathological events, has been complicated by the lack of specific inhibitors and the fact that some of the potent MMPs are multifunctional enzymes. These factors have also hampered the setup of therapeutic strategies targeting MMP activity. A tempting target is the membrane-associated MT1-MMP, which has well-documented importance in matrix degradation but which takes part in more than one pathway in this regard. In this report, we describe the selective targeting of a single function of this enzyme by means of a specific monoclonal antibody against MT1-MMP, raised in an MT1-MMP knock-out mouse. The antibody blocks the enzyme ability to activate proMMP-2 without interfering with the collagenolytic function or the general proteolytic activity of MT1-MMP. Using this antibody, we have shown that the MT1-MMP-catalyzed activation of proMMP-2 is involved in the outgrowth of cultured lymphatic endothelial cells in a collagen matrix in vitro, as well as in lymphatic vessel sprouting assayed ex vivo. This is the first example of the complete inactivation of a single function of a multifunctional MMP and the use of this strategy to pursue its role.
- Published
- 2013
- Full Text
- View/download PDF
41. C4.4A gene ablation is compatible with normal epidermal development and causes modest overt phenotypes
- Author
-
Morten Persson, Michael Ploug, Ivano Di Meo, Birgitte Holst, Benedikte Jacobsen, Andreas Kjaer, Ole Didrik Laerum, Annette Füchtbauer, Lars H. Engelholm, Mette C. Kriegbaum, Ida K. Lund, Gert H. Hansen, Ib Jarle Christensen, Andreas N. Madsen, Ernst-Martin Füchtbauer, and Carsten Friis Rundsten
- Subjects
Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Urinary Bladder ,Biology ,GPI-Linked Proteins ,Article ,Epithelium ,Carcinoma, Lewis Lung ,03 medical and health sciences ,Thinness ,In vivo ,medicine ,Animals ,Regulation of gene expression ,Wound Healing ,Multidisciplinary ,Cell adhesion molecule ,Body Weight ,Gene Expression Regulation, Developmental ,Cancer ,Gene targeting ,medicine.disease ,Magnetic Resonance Imaging ,Water Loss, Insensible ,Phenotype ,Mice, Inbred C57BL ,Urokinase receptor ,030104 developmental biology ,medicine.anatomical_structure ,Gene Targeting ,Female ,Epidermis ,Energy Metabolism ,Tomography, X-Ray Computed ,Cell Adhesion Molecules ,Gene Deletion ,Neoplasm Transplantation ,Subcellular Fractions - Abstract
C4.4A is a modular glycolipid-anchored Ly6/uPAR/α-neurotoxin multidomain protein that exhibits a prominent membrane-associated expression in stratified squamous epithelia. C4.4A is also expressed in various solid cancer lesions, where high expression levels often are correlated to poor prognosis. Circumstantial evidence suggests a role for C4.4A in cell adhesion, migration and invasion, but a well-defined biological function is currently unknown. In the present study, we have generated and characterized the first C4.4A-deficient mouse line to gain insight into the functional significance of C4.4A in normal physiology and cancer progression. The unchallenged C4.4A-deficient mice were viable, fertile, born in a normal Mendelian distribution and, surprisingly, displayed normal development of squamous epithelia. The C4.4A-deficient mice were, nonetheless, significantly lighter than littermate controls predominantly due to differences in fat mass. Congenital C4.4A deficiency delayed migration of keratinocytes enclosing incisional skin wounds in male mice. In chemically induced bladder carcinomas, C4.4A deficiency attenuated the incidence of invasive lesions despite having no effect on total tumour burden. This new C4.4A-deficient mouse line provides a useful platform for future studies on functional aspects of C4.4A in tumour cell invasion in vivo.
- Published
- 2016
- Full Text
- View/download PDF
42. Crystal structures of the ligand-binding region of uPARAP: effect of calcium ion binding
- Author
-
Mingdong Huang, Lars H. Engelholm, Min Liu, Zhipu Luo, Longguang Jiang, Rui Li, Henrik J. Jürgensen, Cai Yuan, and Niels Behrendt
- Subjects
0301 basic medicine ,Models, Molecular ,Conformational change ,Protein Conformation ,Receptors, Cell Surface ,Crystal structure ,Biology ,Crystallography, X-Ray ,Ligands ,Biochemistry ,Extracellular matrix ,03 medical and health sciences ,Humans ,Calcium ion binding ,Molecular Biology ,Binding Sites ,Membrane Glycoproteins ,030102 biochemistry & molecular biology ,Lectin ,Cell Biology ,Urokinase receptor ,Crystallography ,030104 developmental biology ,HEK293 Cells ,Mannose-Binding Lectins ,Biophysics ,Molecular mechanism ,biology.protein ,Calcium ,Mannose receptor - Abstract
The proteins of the mannose receptor (MR) family share a common domain organization and have a broad range of biological functions. Urokinase plasminogen activator receptor-associated protein (uPARAP) (or Endo180) is a member of this family and plays an important role in extracellular matrix remodelling through interaction with its ligands, including collagens and urokinase plasminogen activator receptor (uPAR). We report the crystal structures of the first four domains of uPARAP (also named the ligand-binding region, LBR) at pH 7.4 in Ca2+-bound and Ca2+-free forms. The first domain (cysteine-rich or CysR domain) folds into a new and unique conformation different from the β-trefoil fold of typical CysR domains. The so-called long loop regions (LLRs) of the C-type lectin-like domain (CTLD) 1 and 2 (the third and fourth domain) mediate the direct contacts between these domains. These LLRs undergo a Ca2+-dependent conformational change, and this is likely to be the key structural determinant affecting the overall conformation of uPARAP. Our results provide a molecular mechanism to support the structural flexibility of uPARAP, and shed light on the structural flexibility of other members of the MR family.
- Published
- 2016
43. Targeting a novel bone degradation pathway in primary bone cancer by inactivation of the collagen receptor uPARAP/Endo180
- Author
-
Lars H Engelholm, Maria C Melander, Andreas Hald, Morten Persson, Daniel H Madsen, Henrik J Jürgensen, Kristina Johansson, Christoffer Nielsen, Kirstine S Nørregaard, Signe Z Ingvarsen, Andreas Kjaer, Clement S Trovik, Ole D Laerum, Thomas H Bugge, Johan Eide, Niels Behrendt
- Published
- 2016
- Full Text
- View/download PDF
44. Plasmin‐driven fibrinolysis facilitates skin tumor growth in a gender‐dependent manner
- Author
-
Rasmus Baadsgaard Maerkedahl, Lars H. Engelholm, Kristoffer L. Egerod, Hanne Eickhardt, Leif R. Lund, Andreas Hald, Ole Didrik Laerum, Birgitte Rønø, and Christina Winther Feldborg
- Subjects
Male ,Skin Neoplasms ,Plasmin ,Ovariectomy ,medicine.medical_treatment ,Fibrinogen ,Biochemistry ,Fibrin ,Mice ,Sex Factors ,Fibrinolysis ,Genetics ,medicine ,Animals ,Humans ,Fibrinolysin ,Keratinocyte migration ,Molecular Biology ,Mice, Knockout ,biology ,Chemistry ,Thrombosis ,Neoplasms, Experimental ,medicine.disease ,Extracellular Matrix ,Gene Expression Regulation, Neoplastic ,Cancer research ,biology.protein ,Female ,Tumor necrosis factor alpha ,Laminin ,Skin cancer ,Wound healing ,Biotechnology ,medicine.drug - Abstract
Rearrangement of the skin during wound healing depends on plasmin and plasminogen, which serve to degrade fibrin depositions in the provisional matrix and thereby facilitate keratinocyte migration. In the current study, we investigated whether plasmin and plasminogen likewise played a role during the development of skin cancer. To test this, we set up a chemically induced skin tumor model in a cohort of mice and found that skin tumor growth in Plg(-/-) male mice was reduced by 52% compared with wild-type controls. Histological analyses suggested that the growth-restricting effect of plasminogen deficiency was due to thrombosis and lost patency of the tumor vasculature, resulting in tumor necrosis. The connection between plasmin-dependent fibrinolysis, vascular patency, and tumor growth was further substantiated as the effect of plasminogen deficiency on tumor growth could be reverted by superimposing heterozygous fibrinogen deficiency on Plg(-/-) mice. Tumors derived from these Fib(-/+);Plg(-/-) mice displayed a significantly decreased level of tumor thrombosis compared with Plg(-/-) mice. In summary, these data indicate that plasmin-driven fibrinolysis facilitates tumor growth by maintaining patency of the tumor vasculature.
- Published
- 2012
- Full Text
- View/download PDF
45. Endocytic collagen degradation: a novel mechanism involved in protection against liver fibrosis
- Author
-
Thomas H. Bugge, Henrik J. Jürgensen, Lars H. Engelholm, Ben Vainer, Kristoffer L. Egerod, Signe Ingvarsen, Niels Behrendt, Charlotte A. Madsen, Daniel H. Madsen, Maria C. Melander, Birgitte Rønø, and Andreas Hald
- Subjects
0303 health sciences ,Pathology ,medicine.medical_specialty ,Cirrhosis ,Endocytic cycle ,Matrix metalloproteinase ,Biology ,medicine.disease ,3. Good health ,Pathology and Forensic Medicine ,Collagen receptor ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,030220 oncology & carcinogenesis ,Hepatic stellate cell ,medicine ,Intracellular ,Mannose receptor ,030304 developmental biology - Abstract
Fibrosis of the liver and its end-stage, cirrhosis, represent major health problems worldwide. In these fibrotic conditions, activated fibroblasts and hepatic stellate cells display a net deposition of collagen. This collagen deposition is a major factor leading to liver dysfunction, thus making it crucially important to understand both the collagen synthesis and turnover mechanisms in this condition. Here we show that the endocytic collagen receptor, uPARAP/Endo180, is a major determinant in governing the balance between collagen deposition and degradation. Cirrhotic human livers displayed a marked up-regulation of uPARAP/Endo180 in activated fibroblasts and hepatic stellate cells located close to the collagen deposits. In a hepatic stellate cell line, uPARAP/Endo180 was shown to be active in, and required for, the uptake and intracellular degradation of collagen. To evaluate the functional importance of this collagen receptor in vivo, liver fibrosis was induced in uPARAP/Endo180-deficient mice and littermate wild-type mice by chronic CCl4 administration. A strong up-regulation of uPARAP/Endo180 was observed in wild-type mice, and a quantitative comparison of collagen deposits in the two groups of mice clearly revealed a fibrosis protective role of uPARAP/Endo180. This effect appeared to directly reflect the activity of the collagen receptor, since no compensatory events were noted when comparing the mRNA expression profiles of the two groups of mice in an array system focused on matrix-degrading components. This function of uPARAP/Endo180 defines a novel role of intracellular collagen turnover in fibrosis protection. Copyright © 2012 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
- Published
- 2012
- Full Text
- View/download PDF
46. Conformational Regulation of Urokinase Receptor Function
- Author
-
Henrik Gårdsvoll, Søren Østergaard, Niels Behrendt, Lars H. Engelholm, Michael Ploug, Mette C. Kriegbaum, and Benedikte Jacobsen
- Subjects
Biology ,Biochemistry ,Protein–protein interaction ,03 medical and health sciences ,0302 clinical medicine ,Binding site ,skin and connective tissue diseases ,10. No inequality ,Cell adhesion ,Receptor ,neoplasms ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Cell Biology ,Molecular biology ,biological factors ,Cell biology ,Urokinase receptor ,enzymes and coenzymes (carbohydrates) ,030220 oncology & carcinogenesis ,biology.protein ,Vitronectin ,biological phenomena, cell phenomena, and immunity ,Lamellipodium ,Plasminogen activator - Abstract
The urokinase-type plasminogen activator receptor (uPAR) is a glycolipid-anchored membrane protein with an established role in focalizing uPA-mediated plasminogen activation on cell surfaces. Distinct from this function, uPAR also modulates cell adhesion and migration on vitronectin-rich matrices. Although uPA and vitronectin engage structurally distinct binding sites on uPAR, they nonetheless cooperate functionally, as uPA binding potentiates uPAR-dependent induction of lamellipodia on vitronectin matrices. We now present data advancing the possibility that it is the burial of the β-hairpin in uPA per se into the hydrophobic ligand binding cavity of uPAR that modulates the function of this receptor. Based on these data, we now propose a model in which the inherent interdomain mobility in uPAR plays a major role in modulating its function. Particularly one uPAR conformation, which is stabilized by engagement of the β-hairpin in uPA, favors the proper assembly of an active, compact receptor structure that stimulates lamellipodia induction on vitronectin. This molecular model has wide implications for drug development targeting uPAR function.
- Published
- 2011
- Full Text
- View/download PDF
47. A Novel Functional Role of Collagen Glycosylation
- Author
-
Lars H. Engelholm, Signe Ingvarsen, Niels Behrendt, László Patthy, Daniel H. Madsen, Henrik Gårdsvoll, Maria C. Melander, and Henrik J. Jürgensen
- Subjects
chemistry.chemical_classification ,0303 health sciences ,Glycosylation ,Protein family ,Endocytic cycle ,Cell Biology ,Biology ,Biochemistry ,Collagen receptor ,Extracellular matrix ,Fibronectin type II domain ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,030220 oncology & carcinogenesis ,Glycoprotein ,Molecular Biology ,Mannose receptor ,030304 developmental biology - Abstract
Collagens make up the most abundant component of interstitial extracellular matrices and basement membranes. Collagen remodeling is a crucial process in many normal physiological events and in several pathological conditions. Some collagen subtypes contain specific carbohydrate side chains, the function of which is poorly known. The endocytic collagen receptor urokinase plasminogen activator receptor-associated protein (uPARAP)/Endo180 plays an important role in matrix remodeling through its ability to internalize collagen for lysosomal degradation. uPARAP/Endo180 is a member of the mannose receptor protein family. These proteins all include a fibronectin type II domain and a series of C-type lectin-like domains, of which only a minor part possess carbohydrate recognition activity. At least two of the family members, uPARAP/Endo180 and the mannose receptor, interact with collagens. The molecular basis for this interaction is known to involve the fibronectin type II domain but nothing is known about the function of the lectin domains in this respect. In this study, we have investigated a possible role of the single active lectin domain of uPARAP/Endo180 in the interaction with collagens. By expressing truncated recombinant uPARAP/Endo180 proteins and analyzing their interaction with collagens with high and low levels of glycosylation we demonstrated that this lectin domain interacts directly with glycosylated collagens. This interaction is functionally important because it was found to modulate the endocytic efficiency of the receptor toward highly glycosylated collagens such as basement membrane collagen IV. Surprisingly, this property was not shared by the mannose receptor, which internalized glycosylated collagens independently of its lectin function. This role of modulating its uptake efficiency by a specific receptor is a previously unrecognized function of collagen glycosylation.
- Published
- 2011
- Full Text
- View/download PDF
48. The Non-phagocytic Route of Collagen Uptake
- Author
-
Signe Ingvarsen, Christian Honoré, Niels Behrendt, Charlotte A. Madsen, Thomas H. Bugge, Amanda Moyer, Maria C. Melander, Henrik J. Jürgensen, Peter Garred, Lars H. Engelholm, Sven Burgdorf, Lars Kjøller, and Daniel H. Madsen
- Subjects
0303 health sciences ,media_common.quotation_subject ,Integrin ,Cell Biology ,Biology ,Endocytosis ,Biochemistry ,Collagen receptor ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,medicine ,Receptor ,Fibroblast ,Internalization ,Molecular Biology ,Mannose receptor ,030304 developmental biology ,media_common - Abstract
The degradation of collagens, the most abundant proteins of the extracellular matrix, is involved in numerous physiological and pathological conditions including cancer invasion. An important turnover pathway involves cellular internalization and degradation of large, soluble collagen fragments, generated by initial cleavage of the insoluble collagen fibers. We have previously observed that in primary mouse fibroblasts, this endocytosis of collagen fragments is dependent on the receptor urokinase plasminogen activator receptor-associated protein (uPARAP)/Endo180. Others have identified additional mechanisms of collagen uptake, with different associated receptors, in other cell types. These receptors include β1-integrins, being responsible for collagen phagocytosis, and the mannose receptor. We have now utilized a newly developed monoclonal antibody against uPARAP/Endo180, which down-regulates the receptor protein level on treated cells, to examine the role of uPARAP/Endo180 as a mediator of collagen internalization by a wide range of cultured cell types. With the exception of macrophages, all cells that proved capable of efficient collagen internalization were of mesenchymal origin and all of these utilized uPARAP/Endo180 for their collagen uptake process. Macrophages internalized collagen in a process mediated by the mannose receptor, a protein belonging to the same protein family as uPARAP/Endo180. β1-Integrins were found not to be involved in the endocytosis of soluble collagen, irrespectively of whether this was mediated by uPARAP/Endo180 or the mannose receptor. This further distinguishes these pathways from the phagocytic uptake of particulate collagen.
- Published
- 2011
- Full Text
- View/download PDF
49. Fibroblast Growth Factor Receptor 4 Regulates Tumor Invasion by Coupling Fibroblast Growth Factor Signaling to Extracellular Matrix Degradation
- Author
-
Jouko Lohi, Marko Hyytiäinen, Olli Kallioniemi, Jussi Taipale, Kari Alitalo, Sami Kilpinen, Pipsa Meller, Lars H. Engelholm, Kaisa Lehti, Nami Sugiyama, Signe Ingvarsen, Markku Varjosalo, and Jorma Keski-Oja
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,DNA, Complementary ,Breast Neoplasms ,Mice, SCID ,Adenocarcinoma ,Biology ,Polymerase Chain Reaction ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Growth factor receptor ,Neoplasms ,Internal medicine ,medicine ,Animals ,Homeostasis ,Humans ,Neoplasm Invasiveness ,Receptor, Fibroblast Growth Factor, Type 4 ,Growth factor receptor inhibitor ,Gene Silencing ,RNA, Small Interfering ,030304 developmental biology ,0303 health sciences ,Fibroblast growth factor receptor 2 ,Fibroblast growth factor receptor 1 ,Prostatic Neoplasms ,Fibroblast growth factor receptor 4 ,Fibroblast growth factor receptor 3 ,Extracellular Matrix ,3. Good health ,Fibroblast Growth Factors ,Gene Expression Regulation, Neoplastic ,Endocrinology ,Oncology ,Fibroblast growth factor receptor ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Lymph Nodes ,Cell Division ,Signal Transduction - Abstract
Aberrant expression and polymorphism of fibroblast growth factor receptor 4 (FGFR4) has been linked to tumor progression and anticancer drug resistance. We describe here a novel mechanism of tumor progression by matrix degradation involving epithelial-to-mesenchymal transition in response to membrane-type 1 matrix metalloproteinase (MT1-MMP, MMP-14) induction at the edge of tumors expressing the FGFR4-R388 risk variant. Both FGFR4 and MT1-MMP were upregulated in tissue biopsies from several human cancer types including breast adenocarcinomas, where they were partially coexpressed at the tumor/stroma border and tumor invasion front. The strongest overall coexpression was found in prostate carcinoma. Studies with cultured prostate carcinoma cell lines showed that the FGFR4-R388 variant, which has previously been associated with poor cancer prognosis, increased MT1-MMP–dependent collagen invasion. In this experimental model, knockdown of FGFR4-R388 or MT1-MMP by RNA interference blocked tumor cell invasion and growth in collagen. This was coupled with impaired phosphorylation of FGFR substrate 2 and Src, upregulation of E-cadherin, and suppression of cadherin-11 and N-cadherin. These in vitro results were substantiated by reduced MT1-MMP content and in vivo growth of prostate carcinoma cells after the FGFR4-R388 gene silencing. In contrast, knockdown of the alternative FGFR4-G388 allele enhanced MT1-MMP and invasive tumor cell growth in vivo and within three-dimensional collagen. These results will help to explain the reported association of the FGFR4-R388 variant with the progression and poor prognosis of certain types of tumors. Cancer Res; 70(20); 7851–61. ©2010 AACR.
- Published
- 2010
- Full Text
- View/download PDF
50. Mesenchymal cells reactivate Snail1 expression to drive three-dimensional invasion programs
- Author
-
Thomas L. Saunders, Yuexian Hu, Eric R. Fearon, R. Grant Rowe, Xiao Yan Li, Lars H. Engelholm, Signe Ingvarsen, Ismo Virtanen, Karl-Friedrich Becker, Antonio García de Herreros, Guido T. Bommer, and Stephen J. Weiss
- Subjects
Angiogenesis ,Cellular differentiation ,Regulator ,Gene Expression ,Biology ,Extracellular matrix ,Mesoderm ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Report ,Neoplasms ,Conditional gene knockout ,Animals ,Humans ,Protein Isoforms ,Neoplasm Invasiveness ,Epithelial–mesenchymal transition ,Research Articles ,030304 developmental biology ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,0303 health sciences ,Gene Expression Profiling ,Mesenchymal stem cell ,Cell Differentiation ,Epithelial Cells ,Cell Biology ,Fibroblasts ,Molecular biology ,Cell biology ,030220 oncology & carcinogenesis ,SNAI1 ,Snail Family Transcription Factors ,Transcription Factors - Abstract
Epithelial–mesenchymal transition (EMT) is required for mesodermal differentiation during development. The zinc-finger transcription factor, Snail1, can trigger EMT and is sufficient to transcriptionally reprogram epithelial cells toward a mesenchymal phenotype during neoplasia and fibrosis. Whether Snail1 also regulates the behavior of terminally differentiated mesenchymal cells remains unexplored. Using a Snai1 conditional knockout model, we now identify Snail1 as a regulator of normal mesenchymal cell function. Snail1 expression in normal fibroblasts can be induced by agonists known to promote proliferation and invasion in vivo. When challenged within a tissue-like, three-dimensional extracellular matrix, Snail1-deficient fibroblasts exhibit global alterations in gene expression, which include defects in membrane type-1 matrix metalloproteinase (MT1-MMP)-dependent invasive activity. Snail1-deficient fibroblasts explanted atop the live chick chorioallantoic membrane lack tissue-invasive potential and fail to induce angiogenesis. These findings establish key functions for the EMT regulator Snail1 after terminal differentiation of mesenchymal cells.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.