1
|
Pastorino GA, Sheraj I, Huebner K, Ferrero G, Kunze P, Hartmann A, Hampel C, Husnugil HH, Maiuthed A, Gebhart F, Schlattmann F, Gulec Taskiran AE, Oral G, Palmisano R, Pardini B, Naccarati A, Erlenbach-Wuensch K, Banerjee S, Schneider-Stock R. A partial epithelial-mesenchymal transition signature for highly aggressive colorectal cancer cells that survive under nutrient restriction. J Pathol 2024; 262:347-361. [PMID: 38235615 DOI: 10.1002/path.6240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 10/12/2023] [Accepted: 11/21/2023] [Indexed: 01/19/2024]
Abstract
Partial epithelial-mesenchymal transition (p-EMT) has recently been identified as a hybrid state consisting of cells with both epithelial and mesenchymal characteristics and is associated with the migration, metastasis, and chemoresistance of cancer cells. Here, we describe the induction of p-EMT in starved colorectal cancer (CRC) cells and identify a p-EMT gene signature that can predict prognosis. Functional characterisation of starvation-induced p-EMT in HCT116, DLD1, and HT29 cells showed changes in proliferation, morphology, and drug sensitivity, supported by in vivo studies using the chorioallantoic membrane model. An EMT-specific quantitative polymerase chain reaction (qPCR) array was used to screen for deregulated genes, leading to the establishment of an in silico gene signature that was correlated with poor disease-free survival in CRC patients along with the CRC consensus molecular subtype CMS4. Among the significantly deregulated p-EMT genes, a triple-gene signature consisting of SERPINE1, SOX10, and epidermal growth factor receptor (EGFR) was identified. Starvation-induced p-EMT was characterised by increased migratory potential and chemoresistance, as well as E-cadherin processing and internalisation. Both gene signature and E-cadherin alterations could be reversed by the proteasomal inhibitor MG132. Spatially resolving EGFR expression with high-resolution immunofluorescence imaging identified a proliferation stop in starved CRC cells caused by EGFR internalisation. In conclusion, we have gained insight into a previously undiscovered EMT mechanism that may become relevant when tumour cells are under nutrient stress, as seen in early stages of metastasis. Targeting this process of tumour cell dissemination might help to prevent EMT and overcome drug resistance. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Gil A Pastorino
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ilir Sheraj
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey
| | - Kerstin Huebner
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Giulio Ferrero
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Philipp Kunze
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Chuanpit Hampel
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Arnatchai Maiuthed
- Department of Pharmacology, Mahidol University, Bangkok, Thailand
- Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Florian Gebhart
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fynn Schlattmann
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Aliye Ezgi Gulec Taskiran
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey
- Department of Molecular Biology and Genetics, Baskent University, Ankara, Turkey
| | - Goksu Oral
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey
| | - Ralph Palmisano
- Optical Imaging Competence Centre FAU OICE, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Barbara Pardini
- Italian Institute for Genomic Medicine (IIGM), c/o FPO-IRCCS Candiolo, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Alessio Naccarati
- Italian Institute for Genomic Medicine (IIGM), c/o FPO-IRCCS Candiolo, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Katharina Erlenbach-Wuensch
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sreeparna Banerjee
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkey
- Cancer Systems Biology Laboratory (CanSyl), Orta Dogu Teknik Universitesi, Ankara, Turkey
| | - Regine Schneider-Stock
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
2
|
Yang M, Zhan Y, Hou Z, Wang C, Fan W, Guo T, Li Z, Fang L, Lv S, Li S, Gu C, Ye M, Qin H, Liu Q, Cui X. VLDLR disturbs quiescence of breast cancer stem cells in a ligand-independent function. Front Oncol 2022; 12:887035. [PMID: 36568166 PMCID: PMC9767959 DOI: 10.3389/fonc.2022.887035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer stem cells are responsible for cancer initiation, progression, and drug resistance. However, effective targeting strategies against the cell subpopulation are still limited. Here, we unveil two splice variants of very-low-density lipoprotein receptor, VLDLR-I and -II, which are highly expressed in breast cancer stem cells. In breast cancer cells, VLDLR silencing suppresses sphere formation abilities in vitro and tumor growth in vivo. We find that VLDLR knockdown induces transition from self-renewal to quiescence. Surprisingly, ligand-binding activity is not involved in the cancer-promoting functions of VLDLR-I and -II. Proteomic analysis reveals that citrate cycle and ribosome biogenesis-related proteins are upregulated in VLDLR-I and -II overexpressed cells, suggesting that VLDLR dysregulation is associated with metabolic and anabolic regulation. Moreover, high expression of VLDLR in breast cancer tissues correlates with poor prognosis of patients. Collectively, these findings indicate that VLDLR may be an important therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Mengying Yang
- The First Affiliated Hospital, Dalian Medical University, Dalian, China,Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China,State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Yajing Zhan
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Zhijie Hou
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Chunli Wang
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Wenjun Fan
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Tao Guo
- The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Zhuoshi Li
- The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Lei Fang
- The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shasha Lv
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Sisi Li
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chundong Gu
- The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Mingliang Ye
- Chinese Academy of Sciences (CAS) Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| | - Hongqiang Qin
- Chinese Academy of Sciences (CAS) Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China,*Correspondence: Xiaonan Cui, ; Quentin Liu, ; Hongqiang Qin,
| | - Quentin Liu
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China,State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China,*Correspondence: Xiaonan Cui, ; Quentin Liu, ; Hongqiang Qin,
| | - Xiaonan Cui
- The First Affiliated Hospital, Dalian Medical University, Dalian, China,*Correspondence: Xiaonan Cui, ; Quentin Liu, ; Hongqiang Qin,
| |
Collapse
|
3
|
AGO-RBP crosstalk on target mRNAs: Implications in miRNA-guided gene silencing and cancer. Transl Oncol 2022; 21:101434. [PMID: 35477066 PMCID: PMC9136600 DOI: 10.1016/j.tranon.2022.101434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRNAs) and RNA-binding proteins (RBPs) are important regulators of mRNA translation and stability in eukaryotes. While miRNAs can only bind their target mRNAs in association with Argonaute proteins (AGOs), RBPs directly bind their targets either as single entities or in complex with other RBPs to control mRNA metabolism. miRNA binding in 3' untranslated regions (3' UTRs) of mRNAs facilitates an intricate network of interactions between miRNA-AGO and RBPs, thus determining the fate of overlapping targets. Here, we review the current knowledge on the interplay between miRNA-AGO and multiple RBPs in different cellular contexts, the rules underlying their synergism and antagonism on target mRNAs, as well as highlight the implications of these regulatory modules in cancer initiation and progression.
Collapse
|
4
|
Calvier L, Herz J, Hansmann G. Interplay of Low-Density Lipoprotein Receptors, LRPs, and Lipoproteins in Pulmonary Hypertension. JACC Basic Transl Sci 2022; 7:164-180. [PMID: 35257044 PMCID: PMC8897182 DOI: 10.1016/j.jacbts.2021.09.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 12/21/2022]
Abstract
The low-density lipoprotein receptor (LDLR) gene family includes LDLR, very LDLR, and LDL receptor-related proteins (LRPs) such as LRP1, LRP1b (aka LRP-DIT), LRP2 (aka megalin), LRP4, and LRP5/6, and LRP8 (aka ApoER2). LDLR family members constitute a class of closely related multifunctional, transmembrane receptors, with diverse functions, from embryonic development to cancer, lipid metabolism, and cardiovascular homeostasis. While LDLR family members have been studied extensively in the systemic circulation in the context of atherosclerosis, their roles in pulmonary arterial hypertension (PAH) are understudied and largely unknown. Endothelial dysfunction, tissue infiltration of monocytes, and proliferation of pulmonary artery smooth muscle cells are hallmarks of PAH, leading to vascular remodeling, obliteration, increased pulmonary vascular resistance, heart failure, and death. LDLR family members are entangled with the aforementioned detrimental processes by controlling many pathways that are dysregulated in PAH; these include lipid metabolism and oxidation, but also platelet-derived growth factor, transforming growth factor β1, Wnt, apolipoprotein E, bone morpohogenetic proteins, and peroxisome proliferator-activated receptor gamma. In this paper, we discuss the current knowledge on LDLR family members in PAH. We also review mechanisms and drugs discovered in biological contexts and diseases other than PAH that are likely very relevant in the hypertensive pulmonary vasculature and the future care of patients with PAH or other chronic, progressive, debilitating cardiovascular diseases.
Collapse
Key Words
- ApoE, apolipoprotein E
- Apoer2
- BMP
- BMPR, bone morphogenetic protein receptor
- BMPR2
- COPD, chronic obstructive pulmonary disease
- CTGF, connective tissue growth factor
- HDL, high-density lipoprotein
- KO, knockout
- LDL receptor related protein
- LDL, low-density lipoprotein
- LDLR
- LDLR, low-density lipoprotein receptor
- LRP
- LRP, low-density lipoprotein receptor–related protein
- LRP1
- LRP1B
- LRP2
- LRP4
- LRP5
- LRP6
- LRP8
- MEgf7
- Mesd, mesoderm development
- PAH
- PAH, pulmonary arterial hypertension
- PASMC, pulmonary artery smooth muscle cell
- PDGF
- PDGFR-β, platelet-derived growth factor receptor-β
- PH, pulmonary hypertension
- PPARγ
- PPARγ, peroxisome proliferator-activated receptor gamma
- PVD
- RV, right ventricle/ventricular
- RVHF
- RVSP, right ventricular systolic pressure
- TGF-β1
- TGF-β1, transforming growth factor β1
- TGFBR, transforming growth factor β1 receptor
- TNF, tumor necrosis factor receptor
- VLDLR
- VLDLR, very low density lipoprotein receptor
- VSMC, vascular smooth muscle cell
- Wnt
- apolipoprotein E receptor 2
- endothelial cell
- gp330
- low-density lipoprotein receptor
- mRNA, messenger RNA
- megalin
- monocyte
- multiple epidermal growth factor-like domains 7
- pulmonary arterial hypertension
- pulmonary vascular disease
- right ventricle heart failure
- smooth muscle cell
- very low density lipoprotein receptor
- β-catenin
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
- Pulmonary Vascular Research Center, Hannover Medical School, Hannover, Germany
| |
Collapse
|
5
|
Uhl B, A Mittmann L, Dominik J, Hennel R, Smiljanov B, Haring F, B Schaubächer J, Braun C, Padovan L, Pick R, Canis M, Schulz C, Mack M, Gutjahr E, Sinn P, Heil J, Steiger K, Kanse SM, Weichert W, Sperandio M, Lauber K, Krombach F, Reichel CA. uPA-PAI-1 heteromerization promotes breast cancer progression by attracting tumorigenic neutrophils. EMBO Mol Med 2021; 13:e13110. [PMID: 33998175 PMCID: PMC8185543 DOI: 10.15252/emmm.202013110] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 04/28/2021] [Accepted: 03/25/2021] [Indexed: 01/13/2023] Open
Abstract
High intratumoral levels of urokinase-type plasminogen activator (uPA)-plasminogen activator inhibitor-1 (PAI-1) heteromers predict impaired survival and treatment response in early breast cancer. The pathogenetic role of this protein complex remains obscure. Here, we demonstrate that heteromerization of uPA and PAI-1 multiplies the potential of the single proteins to attract pro-tumorigenic neutrophils. To this end, tumor-released uPA-PAI-1 utilizes very low-density lipoprotein receptor and mitogen-activated protein kinases to initiate a pro-inflammatory program in perivascular macrophages. This enforces neutrophil trafficking to cancerous lesions and skews these immune cells toward a pro-tumorigenic phenotype, thus supporting tumor growth and metastasis. Blockade of uPA-PAI-1 heteromerization by a novel small-molecule inhibitor interfered with these events and effectively prevented tumor progression. Our findings identify a therapeutically targetable, hitherto unknown interplay between hemostasis and innate immunity that drives breast cancer progression. As a personalized immunotherapeutic strategy, blockade of uPA-PAI-1 heteromerization might be particularly beneficial for patients with highly aggressive uPA-PAI-1high tumors.
Collapse
Affiliation(s)
- Bernd Uhl
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Laura A Mittmann
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Julian Dominik
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Roman Hennel
- Department of Radiation OncologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Bojan Smiljanov
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Florian Haring
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Johanna B Schaubächer
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Constanze Braun
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Lena Padovan
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Robert Pick
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Martin Canis
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Christian Schulz
- Department of CardiologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Matthias Mack
- Department of Internal MedicineUniversity of RegensburgRegensburgGermany
| | - Ewgenija Gutjahr
- Institute for PathologyUniversity of HeidelbergHeidelbergGermany
| | - Peter Sinn
- Institute for PathologyUniversity of HeidelbergHeidelbergGermany
| | - Jörg Heil
- Department of Gynecology and ObstetricsUniversity of HeidelbergHeidelbergGermany
| | - Katja Steiger
- Department of PathologyTechnical University of MunichMunichGermany
| | - Sandip M Kanse
- Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Wilko Weichert
- Department of PathologyTechnical University of MunichMunichGermany
- German Cancer Consortium (DKTK), partner site MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Markus Sperandio
- Institute of Cardiovascular Physiology and PathophysiologyLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Kirsten Lauber
- Department of Radiation OncologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Fritz Krombach
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Christoph A Reichel
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| |
Collapse
|
6
|
Expression level of VLDL receptor and VLDL-c levels in the malignant and benign breast tumors: The correlation with miRNA-4465 and miRNA-1297. Mol Cell Probes 2020; 53:101624. [DOI: 10.1016/j.mcp.2020.101624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 01/07/2023]
|
7
|
Abstract
The paradoxical pro-tumorigenic function of plasminogen activator inhibitor 1 (PAI-1, aka Serpin E1) in cancer progression and metastasis has been the subject of an abundant scientific literature that has pointed to a pro-angiogenic role, a growth and migration stimulatory function, and an anti-apoptotic activity, all directed toward promoting tumor growth, cancer cell survival, and metastasis. With uPA, PAI-1 is among the most reliable biomarkers and prognosticators in many cancer types. More recently, a novel pro-tumorigenic function of PAI-1 in cancer-related inflammation has been demonstrated. These multifaceted activities of PAI-1 in cancer progression are explained by the complex structure of PAI-1 and its multiple functions that go beyond its anti-fibrinolytic and anti-plasminogen activation activities. However, despite the multiple evidences supporting a pro-tumorigenic role of PAI-1 in cancer, and the development of several inhibitors, targeting PAI-1, has remained elusive. In this article, the various mechanisms responsible for the pro-tumorigenic functions of PAI-1 are reviewed with emphasis on its more recently described contribution to cancer inflammation. The challenges of targeting PAI-1 in cancer therapy are then discussed.
Collapse
Affiliation(s)
- Marta Helena Kubala
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, University of Southern California, Los Angeles, CA, 90033, USA
- The Saban Research Institute of Children's Hospital, Los Angeles, CA, 90027, USA
| | - Yves Albert DeClerck
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, University of Southern California, Los Angeles, CA, 90033, USA.
- The Saban Research Institute of Children's Hospital, Los Angeles, CA, 90027, USA.
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
8
|
Nakajima K, Tokita Y, Tanaka A, Takahashi S. The VLDL receptor plays a key role in the metabolism of postprandial remnant lipoproteins. Clin Chim Acta 2019; 495:382-393. [PMID: 31078566 DOI: 10.1016/j.cca.2019.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 12/21/2022]
Abstract
A new concept to account for the process of postprandial remnant lipoprotein metabolism is proposed based on the characteristics of lipoprotein particles and their receptors. The characteristics of remnant lipoprotein (RLP) were investigated using an immuno-separation method. The majority of the postprandial lipoproteins increased after fat intake was shown to be VLDL remnants, not chylomicron (CM) remnants, based on the significantly high ratio of apoB100/apoB48 in the RLP and the high degree of similarity in the particle size of the apoB48 and apoB100 carrying lipoproteins, which fluctuate in parallel during a 6 h period after fat intake. The VLDL receptor was discovered as a receptor for TG-rich lipoprotein metabolism and is located in peripheral tissues such as skeletal muscle, adipose tissue, etc., but not in the liver. Postprandial VLDL particles are strongly bound and internalized into cells expressing the VLDL receptor. Ligands that bind to VLDL receptor, such as LPL and Lp(a), present in RLP. The presence of various specific ligands in VLDL remnants may enhance the capacity for binding to the VLDL receptor, which play the role primarily for energy delivery to the peripheral tissues, but is also a causal factor in atherogenic diseases when excessively and/or continuously remained in plasma.
Collapse
Affiliation(s)
- Katsuyuki Nakajima
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan; Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan.
| | - Yoshiharu Tokita
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan; Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan
| | - Akira Tanaka
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan
| | - Sadao Takahashi
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan; Division of Diabetes, Ageo Central General Hospital, Saitama, Japan
| |
Collapse
|
9
|
Valoskova K, Biebl J, Roblek M, Emtenani S, Gyoergy A, Misova M, Ratheesh A, Reis-Rodrigues P, Shkarina K, Larsen ISB, Vakhrushev SY, Clausen H, Siekhaus DE. A conserved major facilitator superfamily member orchestrates a subset of O-glycosylation to aid macrophage tissue invasion. eLife 2019; 8:e41801. [PMID: 30910009 PMCID: PMC6435326 DOI: 10.7554/elife.41801] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/11/2019] [Indexed: 12/29/2022] Open
Abstract
Aberrant display of the truncated core1 O-glycan T-antigen is a common feature of human cancer cells that correlates with metastasis. Here we show that T-antigen in Drosophila melanogaster macrophages is involved in their developmentally programmed tissue invasion. Higher macrophage T-antigen levels require an atypical major facilitator superfamily (MFS) member that we named Minerva which enables macrophage dissemination and invasion. We characterize for the first time the T and Tn glycoform O-glycoproteome of the Drosophila melanogaster embryo, and determine that Minerva increases the presence of T-antigen on proteins in pathways previously linked to cancer, most strongly on the sulfhydryl oxidase Qsox1 which we show is required for macrophage tissue entry. Minerva's vertebrate ortholog, MFSD1, rescues the minerva mutant's migration and T-antigen glycosylation defects. We thus identify a key conserved regulator that orchestrates O-glycosylation on a protein subset to activate a program governing migration steps important for both development and cancer metastasis.
Collapse
Affiliation(s)
| | - Julia Biebl
- Institute of Science and Technology AustriaKlosterneuburgAustria
| | - Marko Roblek
- Institute of Science and Technology AustriaKlosterneuburgAustria
| | - Shamsi Emtenani
- Institute of Science and Technology AustriaKlosterneuburgAustria
| | - Attila Gyoergy
- Institute of Science and Technology AustriaKlosterneuburgAustria
| | - Michaela Misova
- Institute of Science and Technology AustriaKlosterneuburgAustria
| | - Aparna Ratheesh
- Institute of Science and Technology AustriaKlosterneuburgAustria
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical SchoolUniversity of WarwickCoventryUnited Kingdom
| | | | | | - Ida Signe Bohse Larsen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Daria E Siekhaus
- Institute of Science and Technology AustriaKlosterneuburgAustria
| |
Collapse
|
10
|
Brifault C, Gilder AS, Laudati E, Banki M, Gonias SL. Shedding of membrane-associated LDL receptor-related protein-1 from microglia amplifies and sustains neuroinflammation. J Biol Chem 2017; 292:18699-18712. [PMID: 28972143 DOI: 10.1074/jbc.m117.798413] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/22/2017] [Indexed: 12/25/2022] Open
Abstract
In the CNS, microglia are activated in response to injury or infection and in neurodegenerative diseases. The endocytic and cell signaling receptor, LDL receptor-related protein-1 (LRP1), is reported to suppress innate immunity in macrophages and oppose microglial activation. The goal of this study was to identify novel mechanisms by which LRP1 may regulate microglial activation. Using primary cultures of microglia isolated from mouse brains, we demonstrated that LRP1 gene silencing increases expression of proinflammatory mediators; however, the observed response was modest. By contrast, the LRP1 ligand, receptor-associated protein (RAP), robustly activated microglia, and its activity was attenuated in LRP1-deficient cells. An important element of the mechanism by which RAP activated microglia was its ability to cause LRP1 shedding from the plasma membrane. This process eliminated cellular LRP1, which is anti-inflammatory, and generated a soluble product, shed LRP1 (sLRP1), which is potently proinflammatory. Purified sLRP1 induced expression of multiple proinflammatory cytokines and the mRNA encoding inducible nitric-oxide synthase in both LRP1-expressing and -deficient microglia. LPS also stimulated LRP1 shedding, as did the heat-shock protein and LRP1 ligand, calreticulin. Other LRP1 ligands, including α2-macroglobulin and tissue-type plasminogen activator, failed to cause LRP1 shedding. Treatment of microglia with a metalloproteinase inhibitor inhibited LRP1 shedding and significantly attenuated RAP-induced cytokine expression. RAP and sLRP1 both caused neuroinflammation in vivo when administered by stereotaxic injection into mouse spinal cords. Collectively, these results suggest that LRP1 shedding from microglia may amplify and sustain neuroinflammation in response to proinflammatory stimuli.
Collapse
Affiliation(s)
- Coralie Brifault
- From the Department of Pathology, University of California San Diego, La Jolla, California 92093
| | - Andrew S Gilder
- From the Department of Pathology, University of California San Diego, La Jolla, California 92093
| | - Emilia Laudati
- From the Department of Pathology, University of California San Diego, La Jolla, California 92093
| | - Michael Banki
- From the Department of Pathology, University of California San Diego, La Jolla, California 92093
| | - Steven L Gonias
- From the Department of Pathology, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
11
|
Magnussen SN, Hadler-Olsen E, Costea DE, Berg E, Jacobsen CC, Mortensen B, Salo T, Martinez-Zubiaurre I, Winberg JO, Uhlin-Hansen L, Svineng G. Cleavage of the urokinase receptor (uPAR) on oral cancer cells: regulation by transforming growth factor - β1 (TGF-β1) and potential effects on migration and invasion. BMC Cancer 2017; 17:350. [PMID: 28526008 PMCID: PMC5438506 DOI: 10.1186/s12885-017-3349-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 05/12/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Urokinase plasminogen activator (uPA) receptor (uPAR) is up-regulated at the invasive tumour front of human oral squamous cell carcinoma (OSCC), indicating a role for uPAR in tumour progression. We previously observed elevated expression of uPAR at the tumour-stroma interface in a mouse model for OSCC, which was associated with increased proteolytic activity. The tumour microenvironment regulated uPAR expression, as well as its glycosylation and cleavage. Both full-length- and cleaved uPAR (uPAR (II-III)) are involved in highly regulated processes such as cell signalling, proliferation, migration, stem cell mobilization and invasion. The aim of the current study was to analyse tumour associated factors and their effect on uPAR cleavage, and the potential implications for cell proliferation, migration and invasion. METHODS Mouse uPAR was stably overexpressed in the mouse OSCC cell line AT84. The ratio of full-length versus cleaved uPAR as analysed by Western blotting and its regulation was assessed by addition of different protease inhibitors and transforming growth factor - β1 (TGF-β1). The role of uPAR cleavage in cell proliferation and migration was analysed using real-time cell analysis and invasion was assessed using the myoma invasion model. RESULTS We found that when uPAR was overexpressed a proportion of the receptor was cleaved, thus the cells presented both full-length uPAR and uPAR (II-III). Cleavage was mainly performed by serine proteases and urokinase plasminogen activator (uPA) in particular. When the OSCC cells were stimulated with TGF-β1, the production of the uPA inhibitor PAI-1 was increased, resulting in a reduction of uPAR cleavage. By inhibiting cleavage of uPAR, cell migration was reduced, and by inhibiting uPA activity, invasion was reduced. We could also show that medium containing soluble uPAR (suPAR), and cleaved soluble uPAR (suPAR (II-III)), induced migration in OSCC cells with low endogenous levels of uPAR. CONCLUSIONS These results show that soluble factors in the tumour microenvironment, such as TGF-β1, PAI-1 and uPA, can influence the ratio of full length and uPAR (II-III) and thereby potentially effect cell migration and invasion. Resolving how uPAR cleavage is controlled is therefore vital for understanding how OSCC progresses and potentially provides new targets for therapy.
Collapse
Affiliation(s)
- Synnove Norvoll Magnussen
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, N-9037, Tromsø, Norway.
| | - Elin Hadler-Olsen
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, N-9037, Tromsø, Norway.,Diagnostic Clinic - Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| | - Daniela Elena Costea
- Gade Laboratory for Pathology, Department of Clinical Medicine, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Eli Berg
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, N-9037, Tromsø, Norway
| | - Cristiane Cavalcanti Jacobsen
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, N-9037, Tromsø, Norway
| | - Bente Mortensen
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, N-9037, Tromsø, Norway
| | - Tuula Salo
- Cancer and Translational Research Medicine Unit, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital, Oulu, Finland.,Oral and Maxillofacial diseases, Clinicum, University of Helsinki, Helsinki, Finland.,Helsinki University Hospital Helsinki, Helsinki, Finland.,Department of Oral Diagnosis, Oral Pathology Division, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, SP-13414-903, Brazil
| | - Inigo Martinez-Zubiaurre
- Department of Clinical Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Jan-Olof Winberg
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, N-9037, Tromsø, Norway
| | - Lars Uhlin-Hansen
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, N-9037, Tromsø, Norway.,Diagnostic Clinic - Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| | - Gunbjorg Svineng
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, N-9037, Tromsø, Norway
| |
Collapse
|
12
|
Takahashi S. Triglyceride Rich Lipoprotein -LPL-VLDL Receptor and Lp(a)-VLDL Receptor Pathways for Macrophage Foam Cell Formation. J Atheroscler Thromb 2017; 24:552-559. [PMID: 28428482 PMCID: PMC5453679 DOI: 10.5551/jat.rv17004] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Very low-density lipoprotein (VLDL) receptor is a member of the low-density lipoprotein (LDL) receptor family. It binds triglyceride rich lipoprotein (TGRL) but not LDL, because it recognizes apolipoprotein (apo)E only but not apoB. The VLDL receptor functions as a peripheral lipoprotein receptor in concert with lipoprotein lipase (LPL) in heart, muscle, adipose tissue and macrophages. In contrast to the LDL receptor, VLDL receptor binds apo E2/2 VLDL and apoE3/3 VLDL particles, and its expression is not down-regulated by intracellular lipoproteins. It has been reported that both LDL-cholesterol (LDL-C) and postprandial triglyceride (chyromicron and VLDL remnants) are risk factors for human atherosclerotic cardiovascular disease (ASCVD). True ligands such as lipoprotein particles of the VLDL receptor are chyromicron remnant (CMR) and VLDL remnant (postprandial hyperlipidemia). Although the oxidized LDL (oxLDL)-scavenger receptors pathway is considered to be the main mechanism for macrophage foam cell formation, it seems that the TGRL-LPL-VLDL receptor pathway is also involved. Since Lp(a) is one of the ligands for the VLDL receptor, the Lp(a)- VLDL receptor pathway is another potential alternative. The expression of VLDL receptor protein in mouse macrophages is modest compared to that in rabbit and human macrophages, both in vitro and in vivo. Therefore, we need to elucidate the mechanism of human ASCVD not by using the mouse model and scavenger receptors pathway but instead using the rabbit model and VLDL receptor pathway, respectively.
Collapse
|
13
|
Kim BK, Yoo HI, Lee AR, Choi K, Yoon SK. Decreased expression ofVLDLRis inversely correlated with miR-200c in human colorectal cancer. Mol Carcinog 2017; 56:1620-1629. [DOI: 10.1002/mc.22618] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 12/31/2016] [Accepted: 01/20/2017] [Indexed: 02/03/2023]
Affiliation(s)
- Bong-Kyu Kim
- Department of Medical Life Sciences; The Catholic University of Korea; Seoul Korea
| | - Hye-In Yoo
- Department of Medical Life Sciences; The Catholic University of Korea; Seoul Korea
| | - Ah-Reum Lee
- Department of Medical Life Sciences; The Catholic University of Korea; Seoul Korea
| | - Keonwoo Choi
- Department of Medical Life Sciences; The Catholic University of Korea; Seoul Korea
| | - Sungjoo Kim Yoon
- Department of Medical Life Sciences; The Catholic University of Korea; Seoul Korea
| |
Collapse
|
14
|
Appert-Collin A, Bennasroune A, Jeannesson P, Terryn C, Fuhrmann G, Morjani H, Dedieu S. Role of LRP-1 in cancer cell migration in 3-dimensional collagen matrix. Cell Adh Migr 2016; 11:316-326. [PMID: 27463962 DOI: 10.1080/19336918.2016.1215788] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The low-density lipoprotein receptor-related protein-1 (LRP-1) is a member of Low Density Lipoprotein Receptor (LDLR) family, which is ubiquitously expressed and which is described as a multifunctional endocytic receptor which mediates the clearance of various extracellular matrix molecules including serine proteinases, proteinase-inhibitor complexes, and matricellular proteins. Several studies showed that high LRP-1 expression promotes breast cancer cell invasiveness, and LRP-1 invalidation leads to cell motility abrogation in both tumor and non-tumor cells. Furthermore, our group has reported that LRP-1 silencing prevents the invasion of a follicular thyroid carcinoma despite increased pericellular proteolytic activities from MMP2 and uPA using a 2D-cell culture model. As the use of 3D culture systems is becoming more and more popular due to their promise as enhanced models of tissue physiology, the aim of the present work is to characterize for the first time how the 3D collagen type I matrix may impact the ability of LRP-1 to regulate the migratory properties of thyroid carcinoma using as a model FTC-133 cells. Our results show that inhibition of LRP-1 activity or expression leads to morphological changes affecting cell-matrix interactions, reorganizations of the actin-cytoskeleton especially by inhibiting FAK activation and increasing RhoA activity and MLC-2 phosphorylation, thus preventing cell migration. Taken together, our results suggest that LRP-1 silencing leads to a decrease of cell migratory capacity in a 3D configuration.
Collapse
Affiliation(s)
- Aline Appert-Collin
- a Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Unité de Formation et de Recherche Sciences Exactes et Naturelles , Reims , France
| | - Amar Bennasroune
- a Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Unité de Formation et de Recherche Sciences Exactes et Naturelles , Reims , France.,b UMR CNRS 7360, LIEC, Université de Lorraine , Metz , France
| | - Pierre Jeannesson
- c Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Faculté de Pharmacie , Reims , France
| | - Christine Terryn
- d Plateforme d'Imagerie Cellulaire et Tissulaire, URCA , Reims , France
| | - Guy Fuhrmann
- e UMR 7213 CNRS, Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie , Illkirch , France
| | - Hamid Morjani
- c Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Faculté de Pharmacie , Reims , France
| | - Stéphane Dedieu
- a Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims Champagne-Ardenne, Unité de Formation et de Recherche Sciences Exactes et Naturelles , Reims , France
| |
Collapse
|
15
|
Wu J, Strawn TL, Luo M, Wang L, Li R, Ren M, Xia J, Zhang Z, Ma W, Luo T, Lawrence DA, Fay WP. Plasminogen activator inhibitor-1 inhibits angiogenic signaling by uncoupling vascular endothelial growth factor receptor-2-αVβ3 integrin cross talk. Arterioscler Thromb Vasc Biol 2015; 35:111-20. [PMID: 25378411 PMCID: PMC4270947 DOI: 10.1161/atvbaha.114.304554] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Plasminogen activator inhibitor-1 (PAI-1) regulates angiogenesis via effects on extracellular matrix proteolysis and cell adhesion. However, no previous study has implicated PAI-1 in controlling vascular endothelial growth factor (VEGF) signaling. We tested the hypothesis that PAI-1 downregulates VEGF receptor-2 (VEGFR-2) activation by inhibiting a vitronectin-dependent cooperative binding interaction between VEGFR-2 and αVβ3. APPROACH AND RESULTS We studied effects of PAI-1 on VEGF signaling in human umbilical vein endothelial cells. PAI-1 inhibited VEGF-induced phosphorylation of VEGFR-2 in human umbilical vein endothelial cells grown on vitronectin, but not on fibronectin or collagen. PAI-1 inhibited the binding of VEGFR-2 to β3 integrin, VEGFR-2 endocytosis, and intracellular signaling pathways downstream of VEGFR-2. The anti-VEGF effect of PAI-1 was mediated by 2 distinct pathways, one requiring binding to vitronectin and another requiring binding to very low-density lipoprotein receptor. PAI-1 inhibited VEGF-induced angiogenesis in vitro and in vivo, and pharmacological inhibition of PAI-1 promoted collateral arteriole development and recovery of hindlimb perfusion after femoral artery interruption. CONCLUSIONS PAI-1 inhibits activation of VEGFR-2 by VEGF by disrupting a vitronectin-dependent proangiogenic binding interaction involving αVβ3 and VEGFR-2. These results broaden our understanding of the roles of PAI-1, vitronectin, and endocytic receptors in regulating VEGFR-2 activation and suggest novel therapeutic strategies for regulating VEGF signaling.
Collapse
Affiliation(s)
- Jianbo Wu
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.).
| | - Tammy L Strawn
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Mao Luo
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Liqun Wang
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Rong Li
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Meiping Ren
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Jiyi Xia
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Zhuo Zhang
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Weizhong Ma
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Tingting Luo
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Daniel A Lawrence
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - William P Fay
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| |
Collapse
|
16
|
Chee LCY, Hendy J, Purton LE, McArthur GA. ATRA and the specific RARα agonist, NRX195183, have opposing effects on the clonogenicity of pre-leukemic murine AML1-ETO bone marrow cells. Leukemia 2012; 27:1369-80. [PMID: 23228968 DOI: 10.1038/leu.2012.362] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
All-trans retinoic acid (ATRA) is used successfully in the treatment of acute promyelocytic leukemia (APL). ATRA enhances hematopoietic stem cell self-renewal through retinoic acid receptor (RAR)γ activation while promoting differentiation of committed myeloid progenitors through RARα activation. Its lack of success in the treatment of non-APL acute myeloid leukemia (AML) may be related to ATRA's non-selectivity for the RARα and RARγ isotypes, and specific RARα activation may be more beneficial in promoting myeloid differentiation. To investigate this hypothesis, the effects of ATRA and the specific RARα agonist NRX195183 was assessed in AML1-ETO (AE)-expressing murine bone marrow (BM) progenitors. ATRA potentiated the in vitro clonogenicity of these cells while NRX195183 had the opposite effect. Morphological and flow cytometric analysis confirmed a predominantly immature myeloid population in the ATRA-treated AE cells while the NRX195183-treated cells demonstrated an increase in the mature myeloid population. Similarly, NRX195183 treatment promoted myeloid differentiation in an AE9a in vivo murine model. In the ATRA-treated AE cells, gene expression analyses revealed functional networks involving SERPINE1 and bone morphogenetic protein 2; AKT phosphorylation was upregulated. Collectively, these findings confirm the contrasting roles of specific RARα and RARγ activation in the clonogenicity and differentiation of AE cells with potential significant implications in the treatment of non-APL AML using a specific RARα agonist.
Collapse
Affiliation(s)
- L C Y Chee
- Molecular Oncology Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
17
|
Tucker TA, Williams L, Koenig K, Kothari H, Komissarov AA, Florova G, Mazar AP, Allen TC, Bdeir K, Mohan Rao LV, Idell S. Lipoprotein receptor-related protein 1 regulates collagen 1 expression, proteolysis, and migration in human pleural mesothelial cells. Am J Respir Cell Mol Biol 2012; 46:196-206. [PMID: 22298529 DOI: 10.1165/rcmb.2011-0071oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The low-density lipoprotein receptor-related protein 1 (LRP-1) binds and can internalize a diverse group of ligands, including members of the fibrinolytic pathway, urokinase plasminogen activator (uPA), and its receptor, uPAR. In this study, we characterized the role of LRP-1 in uPAR processing, collagen synthesis, proteolysis, and migration in pleural mesothelial cells (PMCs). When PMCs were treated with the proinflammatory cytokines TNF-α and IL-1β, LRP-1 significantly decreased at the mRNA and protein levels (70 and 90%, respectively; P < 0.05). Consequently, uPA-mediated uPAR internalization was reduced by 80% in the presence of TNF-α or IL-1β (P < 0.05). In parallel studies, LRP-1 neutralization with receptor-associated protein (RAP) significantly reduced uPA-dependent uPAR internalization and increased uPAR stability in PMCs. LRP-1-deficient cells demonstrated increased uPAR t(1/2) versus LRP-1-expressing PMCs. uPA enzymatic activity was also increased in LRP-1-deficient and neutralized cells, and RAP potentiated uPA-dependent migration in PMCs. Collagen expression in PMCs was also induced by uPA, and the effect was potentiated in RAP-treated cells. These studies indicate that TNF-α and IL-1β regulate LRP-1 in PMCs and that LRP-1 thereby contributes to a range of pathophysiologically relevant responses of these cells.
Collapse
Affiliation(s)
- Torry A Tucker
- The Texas Lung Injury Institute, The University of Texas Health Science Center at Tyler, 11937 US Highway 271, Biomedical Research Building, Lab C-5, Tyler, TX 75708, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Hypoxia-inducible factor-1 (HIF-1) promotes LDL and VLDL uptake through inducing VLDLR under hypoxia. Biochem J 2012; 441:675-83. [PMID: 21970364 DOI: 10.1042/bj20111377] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Metabolism under hypoxia is significantly different from that under normoxia. It has been well elucidated that HIF-1 (hypoxia-inducible factor-1) plays a central role in regulating glucose metabolism under hypoxia; however, the role of HIF-1 in lipid metabolism has not yet been well addressed. In the present study we demonstrate that HIF-1 promotes LDL (low-density lipoprotein) and VLDL (very-LDL) uptake through regulation of VLDLR (VLDL receptor) gene expression under hypoxia. Increased VLDLR mRNA and protein levels were observed under hypoxic or DFO (deferoxamine mesylate salt) treatment in MCF7, HepG2 and HeLa cells. Using dual-luciferase reporter and ChIP (chromatin immunoprecipitation) assays we confirmed a functional HRE (hypoxia-response element) which is localized at +405 in exon 1 of the VLDLR gene. Knockdown of HIF1A (the α subunit of HIF-1) and VLDLR, but not HIF2A (the α subunit of HIF-2), attenuated hypoxia-induced lipid accumulation through affecting LDL and VLDL uptake. Additionally we also observed a correlation between HIF-1 activity and VLDLR expression in hepatocellular carcinoma specimens. The results of the present study suggest that HIF-1-mediated VLDLR induction influences intracellular lipid accumulation through regulating LDL and VLDL uptake under hypoxia.
Collapse
|
19
|
Rebustini IT, Hayashi T, Reynolds AD, Dillard ML, Carpenter EM, Hoffman MP. miR-200c regulates FGFR-dependent epithelial proliferation via Vldlr during submandibular gland branching morphogenesis. Development 2011; 139:191-202. [PMID: 22115756 DOI: 10.1242/dev.070151] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulation of epithelial proliferation during organ morphogenesis is crucial for normal development, as dysregulation is associated with tumor formation. Non-coding microRNAs (miRNAs), such as miR-200c, are post-transcriptional regulators of genes involved in cancer. However, the role of miR-200c during normal development is unknown. We screened miRNAs expressed in the mouse developing submandibular gland (SMG) and found that miR-200c accumulates in the epithelial end buds. Using both loss- and gain-of-function, we demonstrated that miR-200c reduces epithelial proliferation during SMG morphogenesis. To identify the mechanism, we predicted miR-200c target genes and confirmed their expression during SMG development. We discovered that miR-200c targets the very low density lipoprotein receptor (Vldlr) and its ligand reelin, which unexpectedly regulate FGFR-dependent epithelial proliferation. Thus, we demonstrate that miR-200c influences FGFR-mediated epithelial proliferation during branching morphogenesis via a Vldlr-dependent mechanism. miR-200c and Vldlr may be novel targets for controlling epithelial morphogenesis during glandular repair or regeneration.
Collapse
Affiliation(s)
- Ivan T Rebustini
- Matrix and Morphogenesis Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
20
|
Luo J, Sun X, Gao F, Zhao X, Zhong B, Wang H, Sun Z. Effects of ulinastatin and docetaxel on breast cancer invasion and expression of uPA, uPAR and ERK. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2011; 30:71. [PMID: 21798065 PMCID: PMC3173354 DOI: 10.1186/1756-9966-30-71] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2011] [Accepted: 07/29/2011] [Indexed: 11/23/2022]
Abstract
Objective To investigate the effects of ulinastatin and docetaxel on invasion of breast cancer cells and expression of uPA, uPAR and ERK, breast cancer MDA-MB-231 and MCF-7 cells. Methods The nude mice were treated with PBS, ulinastatin, docetaxel, and ulinastatin plus docetaxel, respectively. Their effects on 1) cell invasion ability was assayed using Transwell; 2) expression of uPA, uPAR and ERK was detected by real time PCR and Western blot; 3) uPA, uPAR and p-ERK protein level in nude mice was quantified by immunohistochemistry. Results 1) Treatment with ulinastatin, docetaxel, and ulinastatin plus docetaxel, respectively, significantly inhibited MDA-MB-231 and MCF-7 cell invasion; 2) mRNA and protein levels of uPA, uPAR and ERK1/2 were inhibited by ulinastatin, but enhanced by docetaxel. Conclusion Ulinastatin can enhance the effects of docetaxel on invasion of breast cancer cells. And that uPA, uPAR and p-ERK expression is obviously inhibited by ulinastatin.
Collapse
Affiliation(s)
- Jie Luo
- Department of Breast, Pancreas, and Thyroid Surgery Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Accelerated urokinase-receptor protein turnover triggered by interference with the addition of the glycolipid anchor. Biochem J 2011; 434:233-42. [PMID: 21143195 DOI: 10.1042/bj20101573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
u-PAR (urokinase-type plasminogen activator receptor), anchored to the cell surface via a glycolipid moiety, drives tumour progression. We previously reported that colon cancer cells (RKO clone 2 FS2), attenuated for in vivo tumorigenicity, are diminished >15-fold for u-PAR display when compared with their tumorigenic isogenic counterparts (RKO clone 2), this disparity not reflecting altered transcription/mRNA stability. FACS, confocal microscopy and Western blotting using a fused u-PAR-EGFP (enhanced green fluorescent protein) cDNA revealed a >14-fold differential in the u-PAR-EGFP signal between the isogenic cells, ruling out alternate splicing as a mechanism. Although metabolic labelling indicated similar synthesis rates, pulse-chase revealed accelerated u-PAR-EGFP turnover in the RKO clone 2 FS2 cells. Expression in RKO clone 2 cells of a u-PAR-EGFP protein unable to accept the glycolipid moiety yielded diminished protein amounts, thus mirroring the low endogenous protein levels evident with RKO clone 2 FS2 cells. Transcript levels for the phosphatidylglycan anchor biosynthesis class B gene required for glycolipid synthesis were reduced by 65% in RKO clone 2 FS2 cells, and forced overexpression in these cells partially restored endogenous u-PAR. Thus attenuated u-PAR levels probably reflects accelerated turnover triggered by inefficient addition of the glycolipid moiety.
Collapse
|
22
|
He L, Lu Y, Wang P, Zhang J, Yin C, Qu S. Up-regulated expression of type II very low density lipoprotein receptor correlates with cancer metastasis and has a potential link to β-catenin in different cancers. BMC Cancer 2010; 10:601. [PMID: 21047397 PMCID: PMC2988033 DOI: 10.1186/1471-2407-10-601] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Accepted: 11/03/2010] [Indexed: 11/24/2022] Open
Abstract
Background Very low density lipoprotein receptor (VLDLR) has been considered as a multiple function receptor due to binding numerous ligands, causing endocytosis and regulating cellular signaling. Our group previously reported that enhanced activity of type II VLDLR (VLDLR II), one subtype of VLDLR, promotes adenocarcinoma SGC7901 cells proliferation and migration. The aim of this study is to explore the expression levels of VLDLR II in human gastric, breast and lung cancer tissues, and to investigate its relationship with clinical characteristics and β-catenin expression status. Methods VLDLR II expression was examined using immunohistochemistry (IHC) and Western blot in tumor tissues from 213 gastric, breast and lung cancer patients, tumor adjacent noncancerous tissues by same methods. Correlations between VLDLR II and clinical features, as well as β-catenin expression status were evaluated by statistical analysis. Results The immunohistochemical staining of VLDLR II showed statistical difference between tumor tissues and tumor adjacent noncancerous tissues in gastric, breast and lung cancers (P = 0.034, 0.018 and 0.043, respectively). Moreover, using Western, we found higher VLDLR II expression levels were associated with lymph node and distant metastasis in gastric and breast cancer (P < 0.05). Furthermore, highly significant positive correlations were found between VLDLR II and β-catenin in gastric cancer (r = 0.689; P < 0.001)breast cancer (r = 0.594; P < 0.001). Conclusions According to the results of the current study, high VLDLR II expression is correlated with lymph node and distant metastasis in gastric and breast cancer patients, the data suggest that VLDLR II may be a clinical marker in cancers, and has a potential link with β-catenin signaling pathway. This is the first to reveal the closer relationship of VLDLR II with clinical information.
Collapse
Affiliation(s)
- Lei He
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | | | | | | | | | | |
Collapse
|
23
|
Gaultier A, Simon G, Niessen S, Dix M, Takimoto S, Cravatt BF, Gonias SL. LDL receptor-related protein 1 regulates the abundance of diverse cell-signaling proteins in the plasma membrane proteome. J Proteome Res 2010; 9:6689-95. [PMID: 20919742 DOI: 10.1021/pr1008288] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
LDL receptor-related protein 1 (LRP1) is an endocytic receptor, reported to regulate the abundance of other receptors in the plasma membrane, including uPAR and tissue factor. The goal of this study was to identify novel plasma membrane proteins, involved in cell-signaling, that are regulated by LRP1. Membrane protein ectodomains were prepared from RAW 264.7 cells in which LRP1 was silenced and control cells using protease K. Peptides were identified by LC-MS/MS. By analysis of spectral counts, 31 transmembrane and secreted proteins were regulated in abundance at least 2-fold when LRP1 was silenced. Validation studies confirmed that semaphorin4D (Sema4D), plexin domain-containing protein-1 (Plxdc1), and neuropilin-1 were more abundant in the membranes of LRP1 gene-silenced cells. Regulation of Plxdc1 by LRP1 was confirmed in CHO cells, as a second model system. Plxdc1 coimmunoprecipitated with LRP1 from extracts of RAW 264.7 cells and mouse liver. Although Sema4D did not coimmunoprecipitate with LRP1, the cell-surface level of Sema4D was increased by RAP, which binds to LRP1 and inhibits binding of other ligands. These studies identify Plxdc1, Sema4D, and neuropilin-1 as novel LRP1-regulated cell-signaling proteins. Overall, LRP1 emerges as a generalized regulator of the plasma membrane proteome.
Collapse
Affiliation(s)
- Alban Gaultier
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, California 92093, United States
| | | | | | | | | | | | | |
Collapse
|
24
|
Di Y, Liu Z, Tian J, Zong Y, Yang P, Qu S. TFPI or uPA-PAI-1 complex affect cell function through expression variation of type II very low density lipoprotein receptor. FEBS Lett 2010; 584:3469-73. [PMID: 20624392 DOI: 10.1016/j.febslet.2010.07.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 07/06/2010] [Accepted: 07/06/2010] [Indexed: 10/19/2022]
Abstract
Very low density lipoprotein receptors (VLDLR) including type I and type II are known to affect cell functions by binding to its extracellular ligands. However, the effect of these ligands on VLDLR expression remains elusive. Tissue factor pathway inhibitor (TFPI) and urokinase plasminogen activator and plasminogen activator inhibitor 1 (uPA-PAI-1) complex, two ligands of VLDLR, were used to examine their effects on VLDLR expression. TFPI treatment decreased type II VLDLR expression, inhibited cell proliferation and migration, and degradated beta-catenin in SGC7901 cells. However, uPA-PAI-1 complex, increased type II VLDLR expression with promoted cell proliferation and migration and stabilization of beta-catenin. These results indicated that extracellular ligands can change the expression of type II VLDLR to affect cell proliferation and migration.
Collapse
Affiliation(s)
- Yong Di
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | |
Collapse
|
25
|
Ueda K, Asai Y, Yoshimura Y, Iwakawa S. Effect of oil-in-water lipid emulsions prepared with fish oil or soybean oil on the growth of MCF-7 cells and HepG2 cells. J Pharm Pharmacol 2010; 60:1069-75. [DOI: 10.1211/jpp.60.8.0013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Abstract
The growth of human breast cancer-derived MCF-7 cells was affected by oil-in-water lipid emulsions prepared with fish oil (FO) rich in n-3 fatty acids (FAs) and egg-yolk phosphatides (EYP) (FO-emulsions), but not by lipid emulsions prepared with soybean oil (SO) and EYP (SO-emulsions). On the other hand, the growth of human hepatocarcinoma HepG2 cells was affected by neither SO-emulsions nor FO-emulsions. The growth inhibition of MCF-7 cells in the presence of FO-emulsions was not affected by trolox, but was inhibited by α-lipoic acid, and was even potentiated by ebselen, which works as an antioxidant as well as a lipoxygenase inhibitor. Since prostaglandin E3, generated from n-3 FAs by cyclooxygenases, has a suppressive effect on tumour cell growth, and increases when lipoxygenases are inhibited, these findings suggest that lipid emulsions incorporating triglycerides of n-3 FAs might be effective in suppressing the growth of MCF-7 cells, possibly via oxidative stress and through eicosanoid production with anti-proliferating activity against cancer cells.
Collapse
Affiliation(s)
- Kumiko Ueda
- Department of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558, Japan
| | - Yukiko Asai
- Department of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558, Japan
| | - Yoshimizu Yoshimura
- Department of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558, Japan
| | - Seigo Iwakawa
- Department of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558, Japan
| |
Collapse
|
26
|
Schneider WJ. Receptor-mediated mechanisms in ovarian follicle and oocyte development. Gen Comp Endocrinol 2009; 163:18-23. [PMID: 19523388 DOI: 10.1016/j.ygcen.2008.11.032] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 11/26/2008] [Accepted: 11/27/2008] [Indexed: 11/21/2022]
Abstract
The normal development of the chicken oocyte within the ovarian follicle depends on the coordinated expression and function of several members of the low density lipoprotein receptor gene family. The human low density lipoprotein receptor (LDLR) is the prototype of the gene family; since its discovery and the elucidation of the medical significance of mutations in the ldlr gene, many additional family members have been discovered and characterized, and some important advances have resulted from studies in the chicken. I describe the analogies as well as the differences that exist between the molecular genetics of the mammalian and avian members of this important gene family, with emphasis on receptor-mediated oocyte growth. Recent progress in the molecular characterization of the chicken genes whose products mediate oocyte growth, follicle development, and accessory pathways is described in detail, and emerging information of preliminary nature is included. As the availability of chicken genome sequence data has enhanced the rate of progress in the field, our understanding of the physiological roles of members of this receptor family in general has already gained from studies in the avian model system.
Collapse
Affiliation(s)
- Wolfgang J Schneider
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Dr. Bohr Gasse 9/2, A-1030 Vienna, Austria.
| |
Collapse
|
27
|
Gaultier A, Hollister M, Reynolds I, Hsieh EH, Gonias SL. LRP1 regulates remodeling of the extracellular matrix by fibroblasts. Matrix Biol 2009; 29:22-30. [PMID: 19699300 DOI: 10.1016/j.matbio.2009.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 08/04/2009] [Accepted: 08/11/2009] [Indexed: 10/25/2022]
Abstract
Low density lipoprotein receptor-related protein (LRP1) is an endocytic receptor for diverse proteases, protease inhibitors, and other plasma membrane proteins, including the urokinase receptor (uPAR). LRP1 also functions in cell-signaling and regulates gene expression. The goal of this study was to determine whether LRP1 regulates remodeling of provisional extracellular matrix (ECM) by fibroblasts. To address this problem, we utilized an in vitro model in which type I collagen was reconstituted and overlaid with fibronectin. Either the collagen or fibronectin was fluorescently-labeled. ECM remodeling by fibroblasts deficient in LRP1, uPAR, or MT1-MMP was studied. MT1-MMP was required for efficient remodeling of the deep collagen layer but not involved in fibronectin remodeling. Instead, fibronectin was remodeled by a system that required urokinase-type plasminogen activator (uPA), uPAR, and exogenously-added plasminogen. LRP1 markedly inhibited fibronectin remodeling by regulating cell-surface uPAR and plasminogen activation. LRP1 also regulated remodeling of the deep collagen layer but not by controlling MT1-MMP. Instead, LRP1 deficiency or inhibition de-repressed a secondary pathway for collagen remodeling, which was active in MT1-MMP-deficient cells but not in uPAR-deficient cells. These results demonstrate that LRP1 regulates ECM remodeling principally by repressing pathways that require plasminogen activation by uPA in association with uPAR.
Collapse
Affiliation(s)
- Alban Gaultier
- Department of Pathology, University of California San Diego, La Jolla, California 92093, USA
| | | | | | | | | |
Collapse
|
28
|
Obesity and breast cancer: the roles of peroxisome proliferator-activated receptor-γ and plasminogen activator inhibitor-1. PPAR Res 2009; 2009:345320. [PMID: 19672469 PMCID: PMC2723729 DOI: 10.1155/2009/345320] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Revised: 05/18/2009] [Accepted: 06/10/2009] [Indexed: 01/01/2023] Open
Abstract
Breast cancer is the most prominent cancer among females in the United States. There are a number of risk factors associated with development of breast cancer, including consumption of a high-fat diet and obesity. Plasminogen activator inhibitor-1 (PAI-1) is a cytokine upregulated in obesity whose expression is correlated with a poor prognosis in breast cancer. As a key mediator of adipogenesis and regulator of adipokine production, peroxisome proliferator-activated receptor-γ (PPAR-γ) is involved in PAI-1 expression from adipose tissue. We summarize the current knowledge linking PPAR-γ and PAI-1 expression to high-fat diet and obesity in the risk of breast cancer.
Collapse
|
29
|
Yang P, Liu Z, Wang H, Tian J, Li Y, Zong Y, Qu S. Enhanced activity of very low density lipoprotein receptor II promotes SGC7901 cell proliferation and migration. Life Sci 2009; 84:402-8. [DOI: 10.1016/j.lfs.2008.12.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2008] [Revised: 11/26/2008] [Accepted: 12/21/2008] [Indexed: 10/21/2022]
|
30
|
Liu Z, Li H, Li Y, Wang Y, Zong Y, Feng Y, Feng Z, Deng Y, Qu S. Up-regulation of VLDL receptor expression and its signaling pathway induced by VLDL and beta-VLDL. ACTA ACUST UNITED AC 2009; 29:1-7. [PMID: 19224153 DOI: 10.1007/s11596-009-0101-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Indexed: 11/26/2022]
Abstract
Very low density lipoprotein receptor (VLDLR) is thought to participate in the pathogenesis of atherosclerosis induced by VLDL and beta-VLDL. The present study was undertaken to elucidate the effects of VLDL and beta-VLDL on VLDLR expression and its signaling pathway. RAW264.7 cells were incubated with VLDL and beta-VLDL. The expression of VLDLR mRNA was detected by RT-PCR. The transcriptional activity of VLDLR gene was detected in recombinant plasmid pGL4.2VR-luciferase transfected RAW264.7. Western blot assay was used to detect the changes of phosphorylated ERK1/2 protein. Inhibitors or activators were used to observe the signal pathway involving VLDLR expression regulation. The results showed that VLDL and beta-VLDL stimulated ERK1/2 activity in a PKC-dependent manner. VLDL or beta-VLDL-induced VLDLR expression on macrophages was extremely abolished by inhibitors ERK1/2 or PKC. Our findings revealed that VLDL or beta-VLDL-induced VLDLR expression via PKC/ERK cascades and the effect was linked to the transcriptional activation of VLDLR gene promoter.
Collapse
Affiliation(s)
- Zhiguo Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Jiang M, Bujo H, Ohwaki K, Unoki H, Yamazaki H, Kanaki T, Shibasaki M, Azuma K, Harigaya K, Schneider WJ, Saito Y. Ang II-stimulated migration of vascular smooth muscle cells is dependent on LR11 in mice. J Clin Invest 2008; 118:2733-46. [PMID: 18618022 DOI: 10.1172/jci32381] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Accepted: 05/21/2008] [Indexed: 11/17/2022] Open
Abstract
Medial-to-intimal migration of SMCs is critical to atherosclerotic plaque formation and remodeling of injured arteries. Considerable amounts of the shed soluble form of the LDL receptor relative LR11 (sLR11) produced by intimal SMCs enhance SMC migration in vitro via upregulation of urokinase-type plasminogen activator receptor (uPAR) expression. Here, we show that circulating sLR11 is a novel marker of carotid intima-media thickness (IMT) and that targeted disruption of the LR11 gene greatly reduces intimal thickening of arteries through attenuation of Ang II-induced migration of SMCs. Serum concentrations of sLR11 were positively correlated with IMT in dyslipidemic subjects, and multivariable regression analysis suggested sLR11 levels as an index of IMT, independent of classical atherosclerosis risk factors. In Lr11-/- mice, femoral artery intimal thickness after cuff placement was decreased, and Ang II-stimulated migration and attachment of SMCs from these mice were largely abolished. In isolated murine SMCs, sLR11 caused membrane ruffle formation via activation of focal adhesion kinase/ERK/Rac1 accompanied by complex formation between uPAR and integrin alphavbeta3, a process accelerated by Ang II. Overproduction of sLR11 decreased the sensitivity of Ang II-induced activation pathways to inhibition by an Ang II type 1 receptor blocker in mice. Thus, we demonstrate a requirement for sLR11 in Ang II-induced SMC migration and propose what we believe is a novel role for sLR11 as a biomarker of carotid IMT.
Collapse
Affiliation(s)
- Meizi Jiang
- Department of Genome Research and Clinical Application, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Tumour expression of the urokinase plasminogen activator correlates with invasive capacity. Consequently, inhibition of this serine protease by physiological inhibitors should decrease invasion and metastasis. However, of the two main urokinase inhibitors, high tumour levels of the type 1 inhibitor actually promote tumour progression, whereas high levels of the type 2 inhibitor decrease tumour growth and metastasis. We propose that the basis of this apparently paradoxical action of two similar serine protease inhibitors lies in key structural differences controlling interactions with components of the extracellular matrix and endocytosis-signalling co-receptors.
Collapse
Affiliation(s)
- David R Croucher
- Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, New South Wales, Australia 2010
| | | | | | | |
Collapse
|
33
|
The plasminogen activator inhibitor "paradox" in cancer. Immunol Lett 2008; 118:116-24. [PMID: 18495253 DOI: 10.1016/j.imlet.2008.03.017] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Revised: 03/29/2008] [Accepted: 03/31/2008] [Indexed: 01/01/2023]
Abstract
Proteolysis in general and specifically the plasminogen activating system regulated by urokinase (uPA) its specific receptor, the GPI membrane anchored urokinase receptor (uPAR) and the specific plasminogen activator inhibitor 1 (PAI-1) plays a major role in tumorigenesis, tumor progression, tumor invasion and metastasis formation. This is exemplified by a body of published work showing a positive correlation between the expression of uPA or uPAR in several tumors and their malignancy. It is generally assumed that such a "pro-malignant" effect of the uPA-uPAR system is mediated by increased local proteolysis thus favoring tumor invasion, by a pro-angiogenic effect of this system and also by uPA-uPAR signaling towards the tumor thereby shifting the tumor phenotype to a more "malignant" one. However, when tumor patients are analyzed for long term survival, those with high levels of the inhibitor of the system, PAI-1 have a much worse prognosis than those with lower PAI-1 levels. This indicates that increased overall proteolysis alone cannot be made responsible for the adverse effects of the plasminogen activating system in tumors. Moreover, it becomes increasingly evident that components of the fibrinolytic system secreted by the tumor cells themselves are not solely responsible for a correlation between the plasminogen activating system and tumor malignancy; components of the plasminogen activating system secreted by stroma cells or cells of the immune system such as macrophages contribute also to the impact of fibrinolysis on malignancy. This review summarizes the evidence for the role of plasminogen activator inhibitor-1 in mediating the malignant phenotype and possible mechanism thereby trying to explain the "PAI-1 paradox in cancer" on a molecular level.
Collapse
|
34
|
Montel V, Gaultier A, Lester RD, Campana WM, Gonias SL. The low-density lipoprotein receptor-related protein regulates cancer cell survival and metastasis development. Cancer Res 2007; 67:9817-24. [PMID: 17942912 DOI: 10.1158/0008-5472.can-07-0683] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Low-density lipoprotein receptor-related protein-1 (LRP-1) is a multifunctional receptor involved in receptor-mediated endocytosis and cell signaling. In this study, we show that LRP-1 is abundantly expressed in severe combined immunodeficient (SCID) mouse xenografts by various human cancer cell lines that express very low or undetectable levels of LRP-1 when cultured in 21% O2 in vitro (standard cell culture conditions). To test whether LRP-1 expression in vivo may be explained by hypoxia in the xenografts, CL16 cells, which are derived from the MDA-MB-435 cell line, were cultured in 1.0% O2. A substantial increase in LRP-1 expression was observed. To test the activity of LRP-1 in cancer progression in vivo, LRP-1 expression was silenced in CL16 cells with short hairpin RNA. These cells formed tumors in SCID mice, in which LRP-1 expression remained silenced. Although LRP-1 gene silencing did not inhibit CL16 cell dissemination from the primary tumors to the lungs, the pulmonary metastases failed to enlarge, suggesting compromised survival or growth at the implantation site. In cell culture experiments, significantly increased cell death was observed when LRP-1-silenced CL16 cells were exposed to CoCl2, which models changes that occur in hypoxia. Furthermore, LRP-1-silenced cells expressed decreased levels of vascular endothelial growth factor in response to 1.0% O2. These results suggest mechanisms by which LRP-1 may facilitate the development and growth of cancer metastases in vivo.
Collapse
Affiliation(s)
- Valérie Montel
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, California 92093-0612, USA
| | | | | | | | | |
Collapse
|
35
|
Croucher D, Saunders D, Stillfried G, Ranson M. A structural basis for differential cell signalling by PAI-1 and PAI-2 in breast cancer cells. Biochem J 2007; 408:203-10. [PMID: 17696882 PMCID: PMC2267350 DOI: 10.1042/bj20070767] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PAI-1 and PAI-2 (plasminogen-activator inibitor types 1 and 2) are inhibitors of cell surface uPA (urokinase plasminogen activator). However, tumour expression of PAI-1 and PAI-2 correlates with poor compared with good patient prognosis in breast cancer respectively. This biological divergence may be related to additional functional roles of PAI-1. For example, the inhibition of uPA by PAI-1 reveals a cryptic high-affinity site within the PAI-1 moiety for the VLDLr (very-low-density-lipoprotein receptor), which sustains cell signalling events initiated by binding of uPA to its receptor. These interactions and subsequent signalling events promote proliferation of breast cancer cells. Biochemical and structural analyses show that, unlike PAI-1, the PAI-2 moiety of uPA-PAI-2 does not contain a high-affinity-binding site for VLDLr, although uPA-PAI-2 is still efficiently endocytosed via this receptor in breast cancer cells. Furthermore, global protein tyrosine phosphorylation events were not sustained by uPA-PAI-2 and cell proliferation was not affected. We thus propose a structurally based mechanism for these differences between PAI-1 and PAI-2 and suggest that PAI-2 is able to inhibit and clear uPA activity without initiating mitogenic signalling events through VLDLr.
Collapse
Affiliation(s)
- David R. Croucher
- *School of Biological Sciences, University of Wollongong, NSW 2522, Australia
| | - Darren N. Saunders
- †Cancer Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- ‡Department of Pathology and Laboratory Medicine, University of British Columbia and Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, BC, Canada V5Z 1L3
| | | | - Marie Ranson
- *School of Biological Sciences, University of Wollongong, NSW 2522, Australia
- To whom correspondence should be addressed (email )
| |
Collapse
|
36
|
Schneider WJ. Low density lipoprotein receptor relatives in chicken ovarian follicle and oocyte development. Cytogenet Genome Res 2007; 117:248-55. [PMID: 17675866 DOI: 10.1159/000103186] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Accepted: 08/08/2006] [Indexed: 11/19/2022] Open
Abstract
The normal development of the chicken oocyte within the ovarian follicle depends on the coordinated expression and function of several members of the low density lipoprotein receptor gene family. The human low density lipoprotein receptor is the prototype of the gene family; since its discovery and the elucidation of the medical significance of mutations in the LDLR gene, many additional family members have been discovered and characterized, and some important advances have resulted from studies in the chicken. I describe the analogies as well as the differences that exist between the molecular genetics of the mammalian and avian members of this important gene family, with emphasis on receptor-mediated oocyte growth. Recent progress in the molecular characterization of the chicken genes whose products mediate oocyte growth, follicle development, and accessory pathways is described in detail, and emerging information of preliminary nature is included. As the availability of chicken genome sequence data has enhanced the rate of progress in the field, our understanding of the physiological roles of members of this receptor family in general has already gained from studies in the avian model system.
Collapse
Affiliation(s)
- W J Schneider
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
37
|
Rolland Y, Demeule M, Michaud-Levesque J, Béliveau R. Inhibition of tumor growth by a truncated and soluble form of melanotransferrin. Exp Cell Res 2007; 313:2910-9. [PMID: 17493610 DOI: 10.1016/j.yexcr.2007.04.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Revised: 04/04/2007] [Accepted: 04/09/2007] [Indexed: 11/20/2022]
Abstract
Melanotransferrin is a glycoprotein expressed at the cell membrane and secreted in the extracellular environment. Recombinant truncated form of membrane-bound melanotransferrin (sMTf) was reported to exert in vitro anti-angiogenic properties. Here we show that sMTf treatment leads to a 50% inhibition of neovascularization in Matrigel implants when stimulated by growth factors. Using a glioblastoma xenograft model, we demonstrate that sMTf delivery at 2.5 and 10 mg/kg/day by micro-osmotic pump inhibits tumor growth by 73% and 91%, respectively. In a lung carcinoma xenograft model, sMTf treatment at 2.5 and 10 mg/kg/day impeded tumor growth by 87% and 97%. Furthermore, subcutaneous glioblastoma and lung carcinoma tumors from mice treated with 10 mg/kg/day of sMTf present insignificant growth toward the study. In association with a reduction in endoglin mRNA expression, the hemoglobin content decreased by half in sMTf-treated glioblastoma tumors. In vitro experiments revealed that NCI-H460 cells treated with sMTf display an inhibition in their invasive capabilities with a concomitant reduction in the expression of the low-density lipoprotein receptor protein and urokinase plasminogen activator receptor. Altogether, our results demonstrate that sMTf exerts anti-cancer and anti-angiogenic activities, suggesting that its administration may provide novel therapeutic strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Yannève Rolland
- Laboratoire de Médecine Moléculaire, Service d'Hémato-Oncologie, Hôpital Ste-Justine - Université du Québec à Montréal, Succursale Centre-ville, Montréal, Québec, Canada
| | | | | | | |
Collapse
|
38
|
Bernstein AM, Twining SS, Warejcka DJ, Tall E, Masur SK. Urokinase receptor cleavage: a crucial step in fibroblast-to-myofibroblast differentiation. Mol Biol Cell 2007; 18:2716-27. [PMID: 17507651 PMCID: PMC1924808 DOI: 10.1091/mbc.e06-10-0912] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2006] [Revised: 04/24/2007] [Accepted: 05/03/2007] [Indexed: 01/20/2023] Open
Abstract
Fibroblasts migrate into and repopulate connective tissue wounds. At the wound edge, fibroblasts differentiate into myofibroblasts, and they promote wound closure. Regulated fibroblast-to-myofibroblast differentiation is critical for regenerative healing. Previous studies have focused on the role in fibroblasts of urokinase plasmingen activator/urokinase plasmingen activator receptor (uPA/uPAR), an extracellular protease system that promotes matrix remodeling, growth factor activation, and cell migration. Whereas fibroblasts have substantial uPA activity and uPAR expression, we discovered that cultured myofibroblasts eventually lost cell surface uPA/uPAR. This led us to investigate the relevance of uPA/uPAR activity to myofibroblast differentiation. We found that fibroblasts expressed increased amounts of full-length cell surface uPAR (D1D2D3) compared with myofibroblasts, which had reduced expression of D1D2D3 but increased expression of the truncated form of uPAR (D2D3) on their cell surface. Retaining full-length uPAR was found to be essential for regulating myofibroblast differentiation, because 1) protease inhibitors that prevented uPAR cleavage also prevented myofibroblast differentiation, and 2) overexpression of cDNA for a noncleavable form of uPAR inhibited myofibroblast differentiation. These data support a novel hypothesis that maintaining full-length uPAR on the cell surface regulates the fibroblast to myofibroblast transition and that down-regulation of uPAR is necessary for myofibroblast differentiation.
Collapse
Affiliation(s)
- Audrey M Bernstein
- Departments of Ophthalmology, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | | | |
Collapse
|
39
|
Ting HJ, Stice JP, Schaff UY, Hui DY, Rutledge JC, Knowlton AA, Passerini AG, Simon SI. Triglyceride-Rich Lipoproteins Prime Aortic Endothelium for an Enhanced Inflammatory Response to Tumor Necrosis Factor-α. Circ Res 2007; 100:381-90. [PMID: 17234968 DOI: 10.1161/01.res.0000258023.76515.a3] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
High levels of triglyceride-rich lipoproteins (TGRLs) in blood are linked to development of atherosclerosis, yet the mechanisms by which these particles initiate inflammation of endothelium are unknown. TGRL isolated from human plasma during the postprandial state was examined for its capacity to bind to cultured human aortic endothelial cells (HAECs) and alter the acute inflammatory response to tumor necrosis factor-α. HAECs were repetitively incubated with dietary levels of freshly isolated TGRL for 2 hours per day for 1 to 3 days to mimic postprandial lipidemia. TGRL induced membrane upregulation of the low-density lipoprotein family receptors LRP and LR11, which was inhibited by the low-density lipoprotein receptor-associated protein-1. TGRLs alone did not elicit inflammation in HAECs but enhanced the inflammatory response via a 10-fold increase in sensitivity to cytokine stimulation. This was reflected by increased mitogen-activated protein kinase activation, nuclear translocation of NF-κB, amplified expression of endothelial selectin and VCAM-1, and a subsequent increase in monocyte-specific recruitment under shear flow as quantified in a microfabricated vascular mimetic device.
Collapse
MESH Headings
- Aorta
- Aortic Diseases/etiology
- Apolipoprotein C-III/metabolism
- Apolipoprotein C-III/pharmacology
- Arteriosclerosis/etiology
- Arteritis/etiology
- Cell Adhesion/drug effects
- Cell Adhesion Molecules/metabolism
- Cells, Cultured/drug effects
- Cells, Cultured/metabolism
- Chylomicrons/blood
- Dietary Fats/administration & dosage
- Dietary Fats/adverse effects
- E-Selectin/biosynthesis
- E-Selectin/genetics
- Endocytosis
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Endothelium, Vascular/cytology
- Fat Emulsions, Intravenous/pharmacology
- Gene Expression Regulation/drug effects
- Humans
- Hypertriglyceridemia/blood
- Hypertriglyceridemia/complications
- Hypoglycemia
- Intercellular Adhesion Molecule-1/biosynthesis
- Intercellular Adhesion Molecule-1/genetics
- LDL-Receptor Related Protein-Associated Protein/pharmacology
- LDL-Receptor Related Proteins/drug effects
- LDL-Receptor Related Proteins/metabolism
- Leukocytes/cytology
- Leukocytes/drug effects
- Lipopolysaccharides/pharmacology
- Lipoproteins, HDL/blood
- Lipoproteins, HDL/toxicity
- Lipoproteins, LDL/blood
- Lipoproteins, LDL/toxicity
- Lipoproteins, VLDL/blood
- Lipoproteins, VLDL/toxicity
- Low Density Lipoprotein Receptor-Related Protein-1/drug effects
- Low Density Lipoprotein Receptor-Related Protein-1/metabolism
- Membrane Transport Proteins/drug effects
- Membrane Transport Proteins/metabolism
- Models, Cardiovascular
- Monocytes/cytology
- Monocytes/drug effects
- NF-kappa B/metabolism
- Oxidative Stress
- Receptors, LDL/drug effects
- Receptors, LDL/metabolism
- Rheology
- Signal Transduction/drug effects
- Triglycerides/blood
- Triglycerides/toxicity
- Tumor Necrosis Factor-alpha/pharmacology
- Tumor Necrosis Factor-alpha/physiology
- Vascular Cell Adhesion Molecule-1/biosynthesis
- Vascular Cell Adhesion Molecule-1/genetics
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Harold J Ting
- Department of Biomedical Engineering, University of California, Davis, Genome and Biomedical Sciences Facility, Davis, CA 95616-5294, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Jiang M, Bujo H, Zhu Y, Yamazaki H, Hirayama S, Kanaki T, Shibasaki M, Takahashi K, Schneider WJ, Saito Y. Pitavastatin attenuates the PDGF-induced LR11/uPA receptor-mediated migration of smooth muscle cells. Biochem Biophys Res Commun 2006; 348:1367-77. [PMID: 16919601 DOI: 10.1016/j.bbrc.2006.07.204] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Accepted: 07/31/2006] [Indexed: 11/26/2022]
Abstract
Statins, inhibitors of HMG-CoA reductase, elicit various actions on vascular cells including the modulation of proliferation and migration of smooth muscle cells (SMCs). Here, we have elucidated the mechanism by which statins, in particular pitavastatin, attenuate the migration activity of SMCs. The expression of LR11, a member of the LDL receptor family and an enhancer of cell surface localization of urokinase-type plasminogen activator receptor (uPAR), is increased in cultured SMCs by treatment with PDGF-BB. Pitavastatin attenuates the PDGF-BB -induced surface expression of LR11 and uPAR. The increased migration of SMCs observed both upon overexpression of LR11 and via stimulation of secretion of soluble LR11 is not reversed by pitavastatin. In vivo studies showed that the SMCs expressing LR11 in plaques are almost congruent with intimal cells expressing nonmuscle myosin heavy chain (SMemb). Pitavastatin reduced the expression of LR11 and SMemb, and the levels of LR11, uPAR, and SMemb in cultured intimal SMCs were reduced to those seen in medial SMCs. We propose that this statin reduces PDGF-induced migration through the attenuation of the LR11/uPAR system in SMCs. Modulation of the LR11/uPAR system with statins suggests a novel treatment strategy for atherogenesis based on suppression of intimal SMC migration.
Collapse
MESH Headings
- Animals
- Becaplermin
- Cell Movement/drug effects
- Cells, Cultured
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- Myosin Heavy Chains/metabolism
- Platelet-Derived Growth Factor/antagonists & inhibitors
- Platelet-Derived Growth Factor/pharmacology
- Proto-Oncogene Proteins c-sis
- Quinolines/pharmacology
- Rabbits
- Receptors, Cell Surface/antagonists & inhibitors
- Receptors, Cell Surface/metabolism
- Receptors, LDL/antagonists & inhibitors
- Receptors, LDL/metabolism
- Receptors, Urokinase Plasminogen Activator
Collapse
Affiliation(s)
- Meizi Jiang
- Department of Genome Research and Clinical Application, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bujo H, Saito Y. Modulation of Smooth Muscle Cell Migration by Members of the Low-Density Lipoprotein Receptor Family. Arterioscler Thromb Vasc Biol 2006; 26:1246-52. [PMID: 16574889 DOI: 10.1161/01.atv.0000219692.78477.17] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Low-density lipoprotein receptor family members (LRs) play a key role in the catabolism of many membrane-associated proteins, such as complexes between proteinases and their receptors, in addition to being involved in lipoprotein metabolism as suspected by the hitherto well-established functions of low-density lipoprotein receptor, in a variety of tissues. Recent studies using receptor-deficient or -overexpressing animals and cells have suggested that certain LRs are important regulators of the migration (and proliferation) of vascular smooth muscle cells (SMCs). LR expression is markedly induced in intimal or medial SMCs during the formation of atherosclerotic lesions. Because LRs can modulate the activity of the urokinase-type plasminogen activator (uPA) receptor and possibly of the platelet-derived growth factor (PDGF) receptor, LRs may influence the migration of SMCs through functional modulation of these membrane receptors. Therefore, SMC migration may be regulated by time-restricted expression of LRs. In agreement with the concept of functional interaction between LRs and membrane signaling receptors, a negative regulator of uPA receptor protein catabolism, LR11, has been identified. Statins modulate the PDGF-induced migration of intimal SMCs via the LR11/uPA receptor cascade. Selective modification of the LRs/uPA receptor/PDGF receptor systems in SMCs may be important for suppression of atherosclerotic plaque formation as well as for preventing intimal thickening after angioplasty.
Collapse
Affiliation(s)
- Hideaki Bujo
- Department of Genome Research and Clinical Application, Chiba University Graduate School of Medicine, Japan.
| | | |
Collapse
|
42
|
Takada H, Imoto I, Tsuda H, Nakanishi Y, Sakakura C, Mitsufuji S, Hirohashi S, Inazawa J. Genomic loss and epigenetic silencing of very-low-density lipoprotein receptor involved in gastric carcinogenesis. Oncogene 2006; 25:6554-62. [PMID: 16715143 DOI: 10.1038/sj.onc.1209657] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Homozygous loss in the genomic sequence, a mechanism for inactivating tumor-suppressor genes (TSGs) in cancer, has been used as a tag for the identification of novel TSGs, and array-based comparative genomic hybridization (array-CGH) has a great potential for high-throughput identification of this change. We identified a homozygous loss of the very-low-density lipoprotein receptor (VLDLR) gene (9p24.2) from genome-wide screening for copy-number alterations in 32 gastric cancer (GC) cell lines using array-CGH. Although previous reports demonstrated mRNA or protein expression of VLDLR in various cancers including GC, the association between genomic losses or epigenetic silencing of this gene and carcinogenesis has never been reported before. Homozygous deletion of VLDLR was also seen in primary GCs, albeit infrequently, and about half of GC cell lines showed lost or reduced VLDLR expression. The VLDLR expression was restored in gene-silenced GC cells after treatment with 5-aza 2'-deoxycytidine. According to methylation analyses, hypermethylation of the VLDLR promoter region, which all of GC lines without its expression showed, occurred in some primary GCs. Restoration of VLDLR type I expression in GC cells reduced colony formation. These results suggest that not only the expression of VLDLR but also genetic or epigenetic silencing of this gene may contribute to tumor formation and be involved in gastric carcinogenesis.
Collapse
Affiliation(s)
- H Takada
- Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Biomedical Science, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Urzúa U, Roby KF, Gangi LM, Cherry JM, Powell JI, Munroe DJ. Transcriptomic analysis of an in vitro murine model of ovarian carcinoma: functional similarity to the human disease and identification of prospective tumoral markers and targets. J Cell Physiol 2006; 206:594-602. [PMID: 16245302 DOI: 10.1002/jcp.20522] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Ovarian cancer is an aggressive disease of poor prognostic when detected at advanced stage. It is widely accepted that the ovarian surface epithelium plays a central role in disease etiology, but little is known about disease progression at the molecular level. To identify genes involved in ovarian tumorigenesis, we carried out a genome-wide transcriptomic analysis of six spontaneously transformed mouse ovarian surface epithelial (MOSE) cell lines, an in vitro model for human ovarian carcinoma. Loess normalization followed by statistical analysis with control of multiple testing resulted in 509 differentially expressed genes using an adjusted P-value < or = 0.05 as cut-off. The top 20 differentially expressed genes included 10 genes (Spp1, Cyp1b1, Btg1, Cfh, Mt1, Mt2, Igfbp5, Gstm1, Gstm2, and Esr1) implicated in various aspects of ovarian carcinomas, and other 3 genes (Gsto1, Lcn7, and Alcam) associated to breast cancer. Upon functional analysis, the majority of alterations affected genes involved in glutathione metabolism and MAPK signaling pathways. Interestingly, over 20% of the aberrantly expressed genes were related to extracellular components, suggestive of potential markers of disease progression. In addition, we identified the genes Pura, Cnn3, Arpc1b, Map4k4, Tgfb1i4, and Crsp2 correlated to in vivo tumorigenic parameters previously reported for these cells. Taken together, our findings support the utility of MOSE cells in studying ovarian cancer biology and as a source of novel diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Ulises Urzúa
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Universidad de Chile, Independencia, Santiago, Chile.
| | | | | | | | | | | |
Collapse
|
44
|
Chaurasia P, Aguirre-Ghiso JA, Liang OD, Gardsvoll H, Ploug M, Ossowski L. A region in urokinase plasminogen receptor domain III controlling a functional association with alpha5beta1 integrin and tumor growth. J Biol Chem 2006; 281:14852-63. [PMID: 16547007 DOI: 10.1074/jbc.m512311200] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Highly expressed urokinase plasminogen activator receptor (uPAR) can interact with alpha5beta1 integrin leading to persistent ERK activation and tumorigenicity. Disrupting this interaction reduces ERK activity, forcing cancer cells into dormancy. We identified a site in uPAR domain III that is indispensable for these effects. A 9-mer peptide derived from a sequence in domain III (residues 240-248) binds purified alpha5beta1 integrin. Substituting a single amino acid (S245A) in this peptide, or in full-length soluble uPAR, impairs binding of the purified integrin. In the recently solved crystal structure of uPAR the Ser-245 is confined to the large external surface of the receptor, a location that is well separated from the central urokinase plasminogen binding cavity. The impact of this site on alpha5beta1 integrin-dependent cell functions was examined by comparing cells induced to express uPAR(wt) or the uPAR(S245A) mutant. Transfecting uPAR(wt) into cells with low endogenous levels of uPAR, inactive integrin, low ERK activity, and a dormant phenotype in vivo restores these functions and reinstates growth in vivo. In contrast, transfection of the same cells with uPAR(S245A) elicits only very small changes. Incubation of highly malignant cells with the wild-type, but not the S245A mutant peptide, disrupts the uPAR integrin interaction leading to down-regulation of ERK activity. The relevance of this binding site, and of the lateral uPAR-alpha5beta1 integrin interaction, to ERK pathway activation and tumor growth implicates it as a possible specific target for cancer therapy.
Collapse
Affiliation(s)
- Pratima Chaurasia
- Department of Medicine, Division of Hematology/Oncology, Mount Sinai School of Medicine, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | | | | | | | | | | |
Collapse
|
45
|
Croucher D, Saunders DN, Ranson M. The urokinase/PAI-2 complex: a new high affinity ligand for the endocytosis receptor low density lipoprotein receptor-related protein. J Biol Chem 2006; 281:10206-13. [PMID: 16459332 DOI: 10.1074/jbc.m513645200] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The efficient inactivation of urokinase plasminogen activator (uPA) by plasminogen activator inhibitor type 2 (PAI-2) at the surface of carcinoma cells is followed by rapid endocytosis of the uPA-PAI-2 complex. We now show that one pathway of this receptor-mediated endocytosis is mediated via the low density lipoprotein receptor-related protein (LRP) in prostate cancer cells. Detailed biochemical analyses using ligand binding assays and surface plasmon resonance revealed a novel and distinct interaction mechanism between native, human LRP and uPA-PAI-2. As reported previously for PAI-1, inhibition of uPA by PAI-2 significantly increased the affinity of the complex for LRP (K(D) of 36 nm for uPA-PAI-2 versus 200 nm for uPA). This interaction was maintained in the presence of uPAR, confirming the validity of this interaction at the cell surface. However, unlike PAI-1, no interaction was observed between LRP and PAI-2 in either the stressed or the relaxed conformation. This suggests that the uPA-PAI-2-LRP interaction is mediated by site(s) within the uPA molecule alone. Thus, as inhibition of uPA by PAI-2 resulted in accelerated clearance of uPA from the cell surface possibly via its increased affinity for LRP, this represents a mechanism through which PAI-2 can clear proteolytic activity from the cell surface. Furthermore, lack of a direct interaction between PAI-2 and LRP implies that downstream signaling events initiated by PAI-1 may not be activated by PAI-2.
Collapse
Affiliation(s)
- David Croucher
- School of Biological Sciences, University of Wollongong, New South Wales 2522
| | | | | |
Collapse
|
46
|
Sid B, Dedieu S, Delorme N, Sartelet H, Rath GM, Bellon G, Martiny L. Human thyroid carcinoma cell invasion is controlled by the low density lipoprotein receptor-related protein-mediated clearance of urokinase plasminogen activator. Int J Biochem Cell Biol 2006; 38:1729-40. [PMID: 16807059 DOI: 10.1016/j.biocel.2006.04.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2006] [Revised: 04/11/2006] [Accepted: 04/12/2006] [Indexed: 11/13/2022]
Abstract
The low density lipoprotein receptor-related protein (LRP), a large scavenger receptor reported to mediate the uptake and degradation of various ligands, emerges as a promising receptor for targeting the invasive behaviour of human cancer cells. However, the accurate function of LRP during tumor invasion seems to be highly dependent on cellular context and remains controversial. The expression patterns of both this receptor and the main proteolytic systems involved in cell invasion were examined in two follicular thyroid carcinoma cell lines exhibiting different invasive phenotypes. We established that a low expression of LRP at the cell surface was associated to elevated extracellular MMP2 and urokinase plasminogen activator (uPA) activities as well as to high invasiveness properties. Surprisingly, neither exogenously added receptor-associated protein, an antagonist of LRP, nor LRP blocking antibodies significantly modified the amount of extracellular MMP2. Furthermore, the invasive phenotype of thyroid carcinoma cells was not related to their matrix metalloproteinases amount since different specific inhibitors of these proteases failed to affect the invasive properties of both cell lines. Additionally, blocking LRP-mediated clearance led to a further increase of the uPA amount and activities and to increased invasiveness in both cell lines. Finally thyroid carcinoma cells aggressiveness was widely increased by exogenous uPA; and anti-uPA antibodies treatments abolished both basal and receptor-associated protein-induced thyroid cell invasion. Overall our results identified the LRP-mediated clearance of uPA as one of the mechanisms involved during the control of human thyroid carcinoma cell invasion.
Collapse
Affiliation(s)
- Brice Sid
- Laboratoire de Biochimie, UMR CNRS 6198, Faculté des Sciences, 51687 Reims, France
| | | | | | | | | | | | | |
Collapse
|
47
|
Chen T, Wu F, Chen FM, Tian J, Qu S. Variations of very low-density lipoprotein receptor subtype expression in gastrointestinal adenocarcinoma cells with various differentiations. World J Gastroenterol 2005; 11:2817-21. [PMID: 15884130 PMCID: PMC4305924 DOI: 10.3748/wjg.v11.i18.2817] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: This study is aimed at investigating the expression and possible significances of very low-density lipoprotein receptor (VLDLR) subtypes in gastroenteric adenocarcinoma tissues and cells with various differentiations.
METHODS: Thirty-one cases of gastroenteric carcinoma/adjacent normal tissues were enrolled in the study, which were diagnosed and classified by the clinicopathological diagnosis. The expression of VLDLR subtypes was detected in gastroenteric carcinoma/adjacent normal tissues and three various differentiated human gastric adenocarcinoma cell lines (MKN28, SGC7901 and MKN45) by reverse transcription polymerase chain reaction (RT-PCR) and Western blot analysis.
RESULTS: Two VLDLR subtypes, namely, type II VLDLR and type I VLDLR, were found to express changes in gastroenteric carcinoma tissues, their adjacent normal tissue, and gastric adenocarcinoma cell lines as well. Type II VLDLR is predominantly expressed in poorly- or moderately-differentiated gastroenteric carcinoma tissues and gastric adenocarcinoma cell lines, whereas type I VLDLR is mainly detected in well-differentiated intestinal carcinoma tissues and gastric adenocarcinoma cells compared with the adjacent normal tissues.
CONCLUSION: The results suggested that the variations of the VLDLR subtype expression might be correlated with the progress and differentiation of gastroenteric carcinoma.
Collapse
Affiliation(s)
- Tao Chen
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | | | | | | | | |
Collapse
|
48
|
Jossin Y. Neuronal migration and the role of reelin during early development of the cerebral cortex. Mol Neurobiol 2005; 30:225-51. [PMID: 15655250 DOI: 10.1385/mn:30:3:225] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2004] [Accepted: 04/29/2004] [Indexed: 11/11/2022]
Abstract
During development, neurons migrate to the cortex radially from periventricular germinative zones as well as tangentially from ganglionic eminences. The vast majority of cortical neurons settle radially in the cortical plate. Neuronal migration requires an exquisite regulation of leading edge extension, nuclear translocation (nucleokinesis), and retraction of trailing processes. During the past few years, several genes and proteins have been identified that are implicated in neuronal migration. Many have been characterized by reference to known mechanisms of neuronal and non-neuronal cell migration in culture; however, probably the most interesting have been identified by gene inactivation or modification in mice and by positional cloning of brain malformation genes in humans and mice. Although it is impossible to provide a fully integrated view, some patterns clearly emerge and are the subject of this article. Specific emphasis is placed on three aspects: first, the role of the actin treadmill, with cyclic formation of filopodial and lamellipodial extensions, in relation to surface events that occur at the leading edge of radially migrating neurons; second, the regulation of microtubule dynamics, which seems to play a key role in nucleokinesis; and third, the mechanisms by which the extracellular protein Reelin regulates neuronal positioning at the end of migration.
Collapse
Affiliation(s)
- Yves Jossin
- Developmental Neurobiology Unit, University of Louvain Medical School, Brussels, Belgium.
| |
Collapse
|
49
|
Raudies O, Kuban RJ, Hamacher F, Klein-Hitpass L, Tchernitsa OI, Sers C, Herzel HP, Schäfer R. Functional analysis and secondary expression profiling of candidate genes deregulated in conjunction with oncogenic Ras signaling. ACTA ACUST UNITED AC 2005; 45:63-84. [PMID: 16023704 DOI: 10.1016/j.advenzreg.2005.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Oliver Raudies
- Laboratory of Molecular Tumor Pathology, Charité, Universitätsmedizin Berlin, D-10117 Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Gliemann J, Hermey G, Nykjaer A, Petersen CM, Jacobsen C, Andreasen PA. The mosaic receptor sorLA/LR11 binds components of the plasminogen-activating system and platelet-derived growth factor-BB similarly to LRP1 (low-density lipoprotein receptor-related protein), but mediates slow internalization of bound ligand. Biochem J 2004; 381:203-12. [PMID: 15053742 PMCID: PMC1133778 DOI: 10.1042/bj20040149] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2004] [Revised: 03/24/2004] [Accepted: 03/30/2004] [Indexed: 11/17/2022]
Abstract
The type-1 receptor sorLA/LR11, a member of the Vps10p-domain receptor family that also contains domains characterizing members of the LDL (low-density lipoprotein) receptor family, has been shown to induce increased uPAR (urokinase receptor) expression as well as enhanced migration and invasion activities in smooth muscle cells in the presence of PDGF-BB (platelet-derived growth factor-BB). Here we show that sorLA interacts with both components of the plasminogen activating system and PDGF-BB similarly to LRP1 (LDL receptor-related protein/alpha2-macroglobulin receptor), which is an important clearance receptor with established functions in controlling uPAR expression as well as PDGF-BB signalling. In contrast with LRP1, sorLA does not interact with alpha2-macroglobulin, which is a binding protein for several growth factors, including PDGF-BB. By using LRP1-deficient cells transfected with sorLA, we demonstrate that sorLA-bound ligand is internalized at a much lower rate than LRP1-bound ligand, and that sorLA is inefficient in regulating cell surface uPAR expression, which depends on rapid internalization of the ternary complex between urokinase-type plasminogen activator, its type-1 inhibitor, and uPAR. Thus, although overlapping with regard to binding profiles, sorLA is substantially less efficient as a clearance receptor than LRP1. We propose that sorLA can divert ligands away from LRP1 and thereby inhibit both their clearance and signalling events mediated by LRP1.
Collapse
Affiliation(s)
- Jørgen Gliemann
- Institute of Medical Biochemistry, University of Aarhus, Ole Worms Allé bldg 170, DK-8000 Arhus C, Denmark.
| | | | | | | | | | | |
Collapse
|