1
|
Alhashmi M, Gremida AME, Maharana SK, Antonaci M, Kerr A, Fu S, Lunn S, Turner DA, Al-Maslamani NA, Liu K, Meschis MM, Sutherland H, Wilson P, Clegg P, Wheeler GN, van 't Hof RJ, Bou-Gharios G, Yamamoto K. Skeletal progenitor LRP1 deficiency causes severe and persistent skeletal defects with Wnt pathway dysregulation. Bone Res 2025; 13:17. [PMID: 39865089 PMCID: PMC11770177 DOI: 10.1038/s41413-024-00393-x] [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: 03/29/2024] [Revised: 09/26/2024] [Accepted: 11/13/2024] [Indexed: 01/28/2025] Open
Abstract
Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional endocytic receptor whose dysfunction is linked to developmental dysplasia of the hip, osteoporosis and osteoarthritis. Our work addresses the critical question of how these skeletal pathologies emerge. Here, we show the abundant expression of LRP1 in skeletal progenitor cells at mouse embryonic stage E10.5 and onwards, especially in the perichondrium, the stem cell layer surrounding developing limbs essential for bone formation. Lrp1 deficiency in these stem cells causes joint fusion, malformation of cartilage/bone template and markedly delayed or lack of primary ossification. These abnormalities, which resemble phenotypes associated with Wnt signalling pathways, result in severe and persistent skeletal defects including a severe deficit in hip joint and patella, and markedly deformed and low-density long bones leading to dwarfism and impaired mobility. Mechanistically, we show that LRP1 regulates core non-canonical Wnt/planar cell polarity (PCP) components that may explain the malformation of long bones. LRP1 directly binds to Wnt5a, facilitates its cell-association and endocytic degradation and recycling. In the developing limbs, LRP1 partially colocalises with Wnt5a and its deficiency alters abundance and distribution of Wnt5a and Vangl2. Finally, using Xenopus as a model system, we show the regulatory role for LRP1 in Wnt/PCP signalling. We propose that in skeletal progenitors, LRP1 plays a critical role in formation and maturity of multiple bones and joints by regulating Wnt signalling, providing novel insights into the fundamental processes of morphogenesis and the emergence of skeletal pathologies.
Collapse
Affiliation(s)
- Mohammad Alhashmi
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulrahman M E Gremida
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Santosh K Maharana
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Marco Antonaci
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Amy Kerr
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Shijian Fu
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Sharna Lunn
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - David A Turner
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Noor A Al-Maslamani
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Ke Liu
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Maria M Meschis
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Hazel Sutherland
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Peter Wilson
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Peter Clegg
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Grant N Wheeler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Robert J van 't Hof
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
- VANTHOF SCIENTIFIC, Torun, Poland
| | - George Bou-Gharios
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK.
| |
Collapse
|
2
|
Harkness JTF, Nayak DA, Sedlacek AL, Cattley R, Hawse WF, Watkins SC, Binder RJ. CD91-mediated reprogramming of DCs by immunogenic heat shock proteins requires the kinases AXL and Fgr. Cell Commun Signal 2024; 22:598. [PMID: 39696592 DOI: 10.1186/s12964-024-01901-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/19/2024] [Indexed: 12/20/2024] Open
Abstract
Immune responses to tumors, comprising adaptive T cells and innate NK cells, arise very early in tumorigeneses and prior to detection of palpable tumors or before tissue pathology is evident. Yet, how nascent tumors evoke dendritic cell maturation and the resulting cytokine responses that are necessary for these effector anti-tumor immune responses is unknown. We have previously shown that CD91 expression on dendritic cells is important for immune surveillance, specifically for generating T cell and NK cell responses to nascent tumors. Here we show that engagement of CD91 by its ligands, the tumor-derived HSPs, triggers intracellular signaling within the dendritic cell and reprograms them to release cytokines and become receptive to other immune mediators. We identify AXL and Fgr as essential adaptor kinases that physically associate with, and phosphorylate, CD91 and are important for transmission of distinct but overlapping signaling in cells. Inhibition of these kinases prevents HSP-induced phosphorylation of signaling cascade components and downstream cytokine production. We show that two different immunogenic HSPs that bind CD91 differentially utilize AXL and Fgr and activate distinct programming of dendritic cells, which is important for the varied immunological responses that tumors evoke. Overall, these findings describe an innate sensing mechanism of nascent tumors by dendritic cells, resulting in initiation of anti-tumor responses via the HSP-CD91 axis.
Collapse
Affiliation(s)
- James Trey F Harkness
- Department of Immunology, 4035 The Assembly, 5051 Centre Ave, Pittsburgh, PA, 15213, USA
| | - Devanshi A Nayak
- Department of Immunology, 4035 The Assembly, 5051 Centre Ave, Pittsburgh, PA, 15213, USA
| | - Abigail L Sedlacek
- Department of Immunology, 4035 The Assembly, 5051 Centre Ave, Pittsburgh, PA, 15213, USA
| | - Richard Cattley
- Department of Immunology, 4035 The Assembly, 5051 Centre Ave, Pittsburgh, PA, 15213, USA
| | - William F Hawse
- Department of Immunology, 4035 The Assembly, 5051 Centre Ave, Pittsburgh, PA, 15213, USA
| | | | - Robert J Binder
- Department of Immunology, 4035 The Assembly, 5051 Centre Ave, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
3
|
Yamamoto K, Scilabra SD, Bonelli S, Jensen A, Scavenius C, Enghild JJ, Strickland DK. Novel insights into the multifaceted and tissue-specific roles of the endocytic receptor LRP1. J Biol Chem 2024; 300:107521. [PMID: 38950861 PMCID: PMC11325810 DOI: 10.1016/j.jbc.2024.107521] [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: 03/13/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
Receptor-mediated endocytosis provides a mechanism for the selective uptake of specific molecules thereby controlling the composition of the extracellular environment and biological processes. The low-density lipoprotein receptor-related protein 1 (LRP1) is a widely expressed endocytic receptor that regulates cellular events by modulating the levels of numerous extracellular molecules via rapid endocytic removal. LRP1 also participates in signalling pathways through this modulation as well as in the interaction with membrane receptors and cytoplasmic adaptor proteins. LRP1 SNPs are associated with several diseases and conditions such as migraines, aortic aneurysms, cardiopulmonary dysfunction, corneal clouding, and bone dysmorphology and mineral density. Studies using Lrp1 KO mice revealed a critical, nonredundant and tissue-specific role of LRP1 in regulating various physiological events. However, exactly how LRP1 functions to regulate so many distinct and specific processes is still not fully clear. Our recent proteomics studies have identified more than 300 secreted proteins that either directly interact with LRP1 or are modulated by LRP1 in various tissues. This review will highlight the remarkable ability of this receptor to regulate secreted molecules in a tissue-specific manner and discuss potential mechanisms underpinning such specificity. Uncovering the depth of these "hidden" specific interactions modulated by LRP1 will provide novel insights into a dynamic and complex extracellular environment that is involved in diverse biological and pathological processes.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.
| | - Simone D Scilabra
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy
| | - Simone Bonelli
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy; Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Anders Jensen
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Lin L, Hu K. Macrophage Function Modulated by tPA Signaling in Mouse Experimental Kidney Disease Models. Int J Mol Sci 2023; 24:11067. [PMID: 37446244 DOI: 10.3390/ijms241311067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Macrophage infiltration and accumulation is a hallmark of chronic kidney disease. Tissue plasminogen activator (tPA) is a serine protease regulating the homeostasis of blood coagulation, fibrinolysis, and matrix degradation, and has been shown to act as a cytokine to trigger various receptor-mediated intracellular signal pathways, modulating macrophage function in response to kidney injury. In this review, we discuss the current understanding of tPA-modulated macrophage function and underlying signaling mechanisms during kidney fibrosis and inflammation.
Collapse
Affiliation(s)
- Ling Lin
- Division of Nephrology, Department of Medicine, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Kebin Hu
- Division of Nephrology, Department of Medicine, Penn State University College of Medicine, Hershey, PA 17033, USA
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
5
|
Montero-Bullón JF, González-Velasco Ó, Isidoro-García M, Lacal J. Integrated in silico MS-based phosphoproteomics and network enrichment analysis of RASopathy proteins. Orphanet J Rare Dis 2021; 16:303. [PMID: 34229750 PMCID: PMC8258961 DOI: 10.1186/s13023-021-01934-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 06/27/2021] [Indexed: 11/30/2022] Open
Abstract
Background RASopathies are a group of syndromes showing clinical overlap caused by mutations in genes affecting the RAS-MAPK pathway. Consequent disruption on cellular signaling leads and is driven by phosphoproteome remodeling. However, we still lack a comprehensive picture of the different key players and altered downstream effectors. Methods An in silico interactome of RASopathy proteins was generated using pathway enrichment analysis/STRING tool, including identification of main hub proteins. We also integrated phosphoproteomic and immunoblotting studies using previous published information on RASopathy proteins and their neighbors in the context of RASopathy syndromes. Data from Phosphosite database (www.phosphosite.org) was collected in order to obtain the potential phosphosites subjected to regulation in the 27 causative RASopathy proteins. We compiled a dataset of dysregulated phosphosites in RASopathies, searched for commonalities between syndromes in harmonized data, and analyzed the role of phosphorylation in the syndromes by the identification of key players between the causative RASopathy proteins and the associated interactome. Results In this study, we provide a curated data set of 27 causative RASopathy genes, identify up to 511 protein–protein associations using pathway enrichment analysis/STRING tool, and identify 12 nodes as main hub proteins. We found that a large group of proteins contain tyrosine residues and their biological processes include but are not limited to the nervous system. Harmonizing published RASopathy phosphoproteomic and immunoblotting studies we identified a total of 147 phosphosites with increased phosphorylation, whereas 47 have reduced phosphorylation. The PKB signaling pathway is the most represented among the dysregulated phosphoproteins within the RASopathy proteins and their neighbors, followed by phosphoproteins implicated in the regulation of cell proliferation and the MAPK pathway. Conclusions This work illustrates the complex network underlying the RASopathies and the potential of phosphoproteomics for dissecting the molecular mechanisms in these syndromes. A combined study of associated genes, their interactome and phosphorylation events in RASopathies, elucidates key players and mechanisms to direct future research, diagnosis and therapeutic windows. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-01934-x.
Collapse
Affiliation(s)
- Javier-Fernando Montero-Bullón
- Metabolic Engineering Group, Department of Microbiology and Genetics, Faculty of Biology, University of Salamanca, 37007, Salamanca, Spain
| | - Óscar González-Velasco
- Bioinformatics and Functional Genomics Group, IBMCC Cancer Research Center, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - María Isidoro-García
- Institute for Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain.,Network for Cooperative Research in Health-RETICS ARADyAL, 37007, Salamanca, Spain.,Department of Clinical Biochemistry, University Hospital of Salamanca, 37007, Salamanca, Spain.,Department of Medicine, University of Salamanca, 37007, Salamanca, Spain
| | - Jesus Lacal
- Institute for Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain. .,Molecular Genetics of Human Diseases Group, Department of Microbiology and Genetics, Faculty of Biology, University of Salamanca, 37007, Salamanca, Spain.
| |
Collapse
|
6
|
Chen J, Su Y, Pi S, Hu B, Mao L. The Dual Role of Low-Density Lipoprotein Receptor-Related Protein 1 in Atherosclerosis. Front Cardiovasc Med 2021; 8:682389. [PMID: 34124208 PMCID: PMC8192809 DOI: 10.3389/fcvm.2021.682389] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/05/2021] [Indexed: 12/26/2022] Open
Abstract
Low-density lipoprotein receptor–related protein-1 (LRP1) is a large endocytic and signaling receptor belonging to the LDL receptor (LDLR) gene family and that is widely expressed in several tissues. LRP1 comprises a large extracellular domain (ECD; 515 kDa, α chain) and a small intracellular domain (ICD; 85 kDa, β chain). The deletion of LRP1 leads to embryonic lethality in mice, revealing a crucial but yet undefined role in embryogenesis and development. LRP1 has been postulated to participate in numerous diverse physiological and pathological processes ranging from plasma lipoprotein homeostasis, atherosclerosis, tumor evolution, and fibrinolysis to neuronal regeneration and survival. Many studies using cultured cells and in vivo animal models have revealed the important roles of LRP1 in vascular remodeling, foam cell biology, inflammation and atherosclerosis. However, its role in atherosclerosis remains controversial. LRP1 not only participates in the removal of atherogenic lipoproteins and proatherogenic ligands in the liver but also mediates the uptake of aggregated LDL to promote the formation of macrophage- and vascular smooth muscle cell (VSMC)-derived foam cells, which causes a prothrombotic transformation of the vascular wall. The dual and opposing roles of LRP1 may also represent an interesting target for atherosclerosis therapeutics. This review highlights the influence of LRP1 during atherosclerosis development, focusing on its dual role in vascular cells and immune cells.
Collapse
Affiliation(s)
- Jiefang Chen
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Su
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shulan Pi
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Mao
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
From the low-density lipoprotein receptor-related protein 1 to neuropathic pain: a potentially novel target. Pain Rep 2021; 6:e898. [PMID: 33981930 PMCID: PMC8108589 DOI: 10.1097/pr9.0000000000000898] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/21/2020] [Accepted: 12/25/2020] [Indexed: 12/12/2022] Open
Abstract
The low-density lipoprotein receptor–related protein 1 plays a major role in the regulation of neuroinflammation, neurodegeneration, neuroregeneration, neuropathic pain, and deficient cognitive functions. This review describes the roles of the low-density lipoprotein receptor–related protein 1 (LRP-1) in inflammatory pathways, nerve nerve degeneration and -regeneration and in neuropathic pain. Induction of LRP-1 is able to reduce the activation of the proinflammatory NFκB-mediated pathway and the mitogen-activated protein kinase (MAPK) c-Jun N-terminal kinase and p38 signaling pathways, in turn decreasing the production of inflammatory mediators. Low-density lipoprotein receptor-related protein 1 activation also decreases reactive astrogliosis and polarizes microglial cells and macrophages from a proinflammatory phenotype (M1) to an anti-inflammatory phenotype (M2), attenuating the neuroinflammatory environment. Low-density lipoprotein receptor-related protein 1 can also modulate the permeability of the blood–brain barrier and the blood–nerve barrier, thus regulating the infiltration of systemic insults and cells into the central and the peripheral nervous system, respectively. Furthermore, LRP-1 is involved in the maturation of oligodendrocytes and in the activation, migration, and repair phenotype of Schwann cells, therefore suggesting a major role in restoring the myelin sheaths upon injury. Low-density lipoprotein receptor-related protein 1 activation can indirectly decrease neurodegeneration and neuropathic pain by attenuation of the inflammatory environment. Moreover, LRP-1 agonists can directly promote neural cell survival and neurite sprouting, decrease cell death, and attenuate pain and neurological disorders by the inhibition of MAPK c-Jun N-terminal kinase and p38-pathway and activation of MAPK extracellular signal–regulated kinase pathway. In addition, activation of LRP-1 resulted in better outcomes for neuropathies such as Alzheimer disease, nerve injury, or diabetic peripheral neuropathy, attenuating neuropathic pain and improving cognitive functions. To summarize, LRP-1 plays an important role in the development of different experimental diseases of the nervous system, and it is emerging as a very interesting therapeutic target.
Collapse
|
8
|
Au DT, Arai AL, Fondrie WE, Muratoglu SC, Strickland DK. Role of the LDL Receptor-Related Protein 1 in Regulating Protease Activity and Signaling Pathways in the Vasculature. Curr Drug Targets 2019; 19:1276-1288. [PMID: 29749311 DOI: 10.2174/1389450119666180511162048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 04/24/2018] [Accepted: 04/24/2018] [Indexed: 12/22/2022]
Abstract
Aortic aneurysms represent a significant clinical problem as they largely go undetected until a rupture occurs. Currently, an understanding of mechanisms leading to aneurysm formation is limited. Numerous studies clearly indicate that vascular smooth muscle cells play a major role in the development and response of the vasculature to hemodynamic changes and defects in these responses can lead to aneurysm formation. The LDL receptor-related protein 1 (LRP1) is major smooth muscle cell receptor that has the capacity to mediate the endocytosis of numerous ligands and to initiate and regulate signaling pathways. Genetic evidence in humans and mouse models reveal a critical role for LRP1 in maintaining the integrity of the vasculature. Understanding the mechanisms by which this is accomplished represents an important area of research, and likely involves LRP1's ability to regulate levels of proteases known to degrade the extracellular matrix as well as its ability to modulate signaling events.
Collapse
Affiliation(s)
- Dianaly T Au
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States
| | - Allison L Arai
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States
| | - William E Fondrie
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States
| | - Selen C Muratoglu
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, MD, United States
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, MD, United States.,Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, MD, United States
| |
Collapse
|
9
|
Bres EE, Faissner A. Low Density Receptor-Related Protein 1 Interactions With the Extracellular Matrix: More Than Meets the Eye. Front Cell Dev Biol 2019; 7:31. [PMID: 30931303 PMCID: PMC6428713 DOI: 10.3389/fcell.2019.00031] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/25/2019] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a biological substrate composed of collagens, proteoglycans and glycoproteins that ensures proper cell migration and adhesion and keeps the cell architecture intact. The regulation of the ECM composition is a vital process strictly controlled by, among others, proteases, growth factors and adhesion receptors. As it appears, ECM remodeling is also essential for proper neuronal and glial development and the establishment of adequate synaptic signaling. Hence, disturbances in ECM functioning are often present in neurodegenerative diseases like Alzheimer’s disease. Moreover, mutations in ECM molecules are found in some forms of epilepsy and malfunctioning of ECM-related genes and pathways can be seen in, for example, cancer or ischemic injury. Low density lipoprotein receptor-related protein 1 (Lrp1) is a member of the low density lipoprotein receptor family. Lrp1 is involved not only in ligand uptake, receptor mediated endocytosis and lipoprotein transport—functions shared by low density lipoprotein receptor family members—but also regulates cell surface protease activity, controls cellular entry and binding of toxins and viruses, protects against atherosclerosis and acts on many cell signaling pathways. Given the plethora of functions, it is not surprising that Lrp1 also impacts the ECM and is involved in its remodeling. This review focuses on the role of Lrp1 and some of its major ligands on ECM function. Specifically, interactions with two Lrp1 ligands, integrins and tissue plasminogen activator are described in more detail.
Collapse
Affiliation(s)
- Ewa E Bres
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
10
|
Potere N, Del Buono MG, Niccoli G, Crea F, Toldo S, Abbate A. Developing LRP1 Agonists into a Therapeutic Strategy in Acute Myocardial Infarction. Int J Mol Sci 2019; 20:E544. [PMID: 30696029 PMCID: PMC6387161 DOI: 10.3390/ijms20030544] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/14/2019] [Accepted: 01/25/2019] [Indexed: 12/16/2022] Open
Abstract
Cardioprotection refers to a strategy aimed at enhancing survival pathways in the injured yet salvageable myocardium following ischemia-reperfusion. Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional receptor that can be targeted following reperfusion, to induce a cardioprotective signaling through the activation of the reperfusion injury salvage kinase (RISK) pathway. The data from preclinical studies with non-selective and selective LRP1 agonists are promising, showing a large therapeutic window for intervention to reduce infarct size after ischemia-reperfusion. A pilot clinical trial with plasma derived α1-antitrypsin (AAT), a naturally occurring LRP1 agonist, supports the translational value of LRP1 as a novel therapeutic target for cardioprotection. A phase I study with a selective LRP1 agonist has been completed showing no toxicity. These findings may open the way to early phase clinical studies with pharmacologic LRP1 activation in patients with acute myocardial infarction (AMI).
Collapse
Affiliation(s)
- Nicola Potere
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
- Unit of Cardiovascular Sciences, Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy.
| | - Marco Giuseppe Del Buono
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Giampaolo Niccoli
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Stefano Toldo
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
11
|
Wujak L, Schnieder J, Schaefer L, Wygrecka M. LRP1: A chameleon receptor of lung inflammation and repair. Matrix Biol 2017; 68-69:366-381. [PMID: 29262309 DOI: 10.1016/j.matbio.2017.12.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 12/17/2022]
Abstract
The lung displays a remarkable capability to regenerate following injury. Considerable effort has been made thus far to understand the cardinal processes underpinning inflammation and reconstruction of lung tissue. However, the factors determining the resolution or persistence of inflammation and efficient wound healing or aberrant remodeling remain largely unknown. Low density lipoprotein receptor-related protein 1 (LRP1) is an endocytic/signaling cell surface receptor which controls cellular and molecular mechanisms driving the physiological and pathological inflammatory reactions and tissue remodeling in several organs. In this review, we will discuss the impact of LRP1 on the consecutive steps of the inflammatory response and its role in the balanced tissue repair and aberrant remodeling in the lung.
Collapse
Affiliation(s)
- Lukasz Wujak
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Jennifer Schnieder
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Liliana Schaefer
- Goethe University School of Medicine, University Hospital, Theodor-Stern Kai 7, 60590 Frankfurt am Main, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany; Member of the German Center for Lung Research (DZL), Germany.
| |
Collapse
|
12
|
Xian X, Ding Y, Dieckmann M, Zhou L, Plattner F, Liu M, Parks JS, Hammer RE, Boucher P, Tsai S, Herz J. LRP1 integrates murine macrophage cholesterol homeostasis and inflammatory responses in atherosclerosis. eLife 2017; 6:e29292. [PMID: 29144234 PMCID: PMC5690284 DOI: 10.7554/elife.29292] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 10/22/2017] [Indexed: 12/11/2022] Open
Abstract
Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional cell surface receptor with diverse physiological roles, ranging from cellular uptake of lipoproteins and other cargo by endocytosis to sensor of the extracellular environment and integrator of a wide range of signaling mechanisms. As a chylomicron remnant receptor, LRP1 controls systemic lipid metabolism in concert with the LDL receptor in the liver, whereas in smooth muscle cells (SMC) LRP1 functions as a co-receptor for TGFβ and PDGFRβ in reverse cholesterol transport and the maintenance of vascular wall integrity. Here we used a knockin mouse model to uncover a novel atheroprotective role for LRP1 in macrophages where tyrosine phosphorylation of an NPxY motif in its intracellular domain initiates a signaling cascade along an LRP1/SHC1/PI3K/AKT/PPARγ/LXR axis to regulate and integrate cellular cholesterol homeostasis through the expression of the major cholesterol exporter ABCA1 with apoptotic cell removal and inflammatory responses.
Collapse
Affiliation(s)
- Xunde Xian
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
| | - Yinyuan Ding
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
- Key Laboratory of Medical Electrophysiology, Ministry of Education of ChinaInstitute of Cardiovascular Research, Southwest Medical UniversityLuzhouChina
| | - Marco Dieckmann
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
| | - Li Zhou
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
| | - Florian Plattner
- Department of PsychiatryUniversity of Texas Southwestern Medical CenterDallasUnited States
- Center for Translational Neurodegeneration ResearchUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Mingxia Liu
- Section on Molecular Medicine, Department of Internal MedicineWake Forest School of MedicineWinston-SalemNorth Carolina
| | - John S Parks
- Section on Molecular Medicine, Department of Internal MedicineWake Forest School of MedicineWinston-SalemNorth Carolina
| | - Robert E Hammer
- Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasUnited States
| | | | - Shirling Tsai
- Department of SurgeryUT Southwestern Medical CenterDallasUnited States
- Dallas VA Medical CenterDallasUnited States
| | - Joachim Herz
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
- Center for Translational Neurodegeneration ResearchUniversity of Texas Southwestern Medical CenterDallasUnited States
- Department of NeuroscienceUT SouthwesternDallasUnited States
- Department of Neurology and NeurotherapeuticsUT SouthwesternDallasUnited States
| |
Collapse
|
13
|
Yang T, Williams BO. Low-Density Lipoprotein Receptor-Related Proteins in Skeletal Development and Disease. Physiol Rev 2017; 97:1211-1228. [PMID: 28615463 DOI: 10.1152/physrev.00013.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 03/07/2017] [Accepted: 03/15/2017] [Indexed: 02/06/2023] Open
Abstract
The identification of the low-density lipoprotein receptor (LDLR) provided a foundation for subsequent studies in lipoprotein metabolism, receptor-mediated endocytosis, and many other fundamental biological functions. The importance of the LDLR led to numerous studies that identified homologous molecules and ultimately resulted in the description of the LDL-receptor superfamily, a group of proteins that contain domains also found in the LDLR. Subsequent studies have revealed that members of the LDLR-related protein family play roles in regulating many aspects of signal transduction. This review is focused on the roles of selected members of this protein family in skeletal development and disease. We present background on the identification of this subgroup of receptors, discuss the phenotypes associated with alterations in their function in human patients and mouse models, and describe the current efforts to therapeutically target these proteins to treat human skeletal disease.
Collapse
Affiliation(s)
- Tao Yang
- Program in Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Bart O Williams
- Program in Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| |
Collapse
|
14
|
van der Meulen T, Swarts S, Fischer W, van der Geer P. Identification of STS-1 as a novel ShcA-binding protein. Biochem Biophys Res Commun 2017; 490:1334-1339. [PMID: 28690151 DOI: 10.1016/j.bbrc.2017.07.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/05/2017] [Indexed: 11/19/2022]
Abstract
ShcA is a cytoplasmic signaling protein that supports signal transduction by receptor protein-tyrosine kinases by providing auxiliary tyrosine phosphorylation sites that engage additional signaling proteins. The principal binding partner for tyrosine phosphorylation sites on ShcA is Grb2. In the current study, we have used phosphotyrosine-containing peptides to isolate and identify STS-1 as a novel ShcA-binding protein. Our results further show that the interaction between STS-1 and ShcA is regulated in response to EGF receptor activation.
Collapse
Affiliation(s)
- Talitha van der Meulen
- Department of Chemistry and Biochemistry, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182-1030, USA
| | - Spencer Swarts
- Department of Chemistry and Biochemistry, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182-1030, USA
| | - Wolfgang Fischer
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute, 10010 North Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Peter van der Geer
- Department of Chemistry and Biochemistry, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182-1030, USA.
| |
Collapse
|
15
|
Dissmore T, Seye CI, Medeiros DM, Weisman GA, Bradford B, Mamedova L. The P2Y2 receptor mediates uptake of matrix-retained and aggregated low density lipoprotein in primary vascular smooth muscle cells. Atherosclerosis 2016; 252:128-135. [PMID: 27522265 PMCID: PMC5060008 DOI: 10.1016/j.atherosclerosis.2016.07.927] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/25/2016] [Accepted: 07/27/2016] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND AIMS The internalization of aggregated low-density lipoproteins (agLDL) mediated by low-density lipoprotein receptor related protein (LRP1) may involve the actin cytoskeleton in ways that differ from the endocytosis of soluble LDL by the LDL receptor (LDLR). This study aims to define novel mechanisms of agLDL uptake through modulation of the actin cytoskeleton, to identify molecular targets involved in foam cell formation in vascular smooth muscle cells (VSMCs). The critical observation that formed the basis for these studies is that under pathophysiological conditions, nucleotide release from blood-derived and vascular cells activates SMC P2Y2 receptors (P2Y2Rs) leading to rearrangement of the actin cytoskeleton and cell motility. Therefore, we tested the hypothesis that P2Y2R activation mediates agLDL uptake by VSMCs. METHODS Primary VSMCs were isolated from aortas of wild type (WT) C57BL/6 and.P2Y2R-/- mice to investigate whether P2Y2R activation modulates LRP1 expression. Cells were transiently transfected with cDNA encoding a hemagglutinin-tagged (HA-tagged) WT P2Y2R, or a mutant P2Y2R that unlike the WT P2Y2R does not bind the cytoskeletal actin-binding protein filamin-A (FLN-A). RESULTS P2Y2R activation significantly increased agLDL uptake, and LRP1 mRNA expression decreased in P2Y2R-/- VSMCs versus WT. SMCs, expressing P2Y2R defective in FLN-A binding, exhibit 3-fold lower LDLR expression levels than SMCs expressing WT P2Y2R, while cells transfected with WT P2Y2R show greater agLDL uptake in both WT and P2Y2R-/- VSMCs versus cells transfected with the mutant P2Y2R. CONCLUSIONS Together, these results show that both LRP1 and LDLR expression and agLDL uptake are regulated by P2Y2R in VSMCs, and that agLDL uptake due to P2Y2R activation is dependent upon cytoskeletal reorganization mediated by P2Y2R binding to FLN-A.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- Aorta/metabolism
- Cell Movement
- Cells, Cultured
- Cytoskeleton/metabolism
- Dose-Response Relationship, Drug
- Endocytosis
- Filamins/metabolism
- Foam Cells/metabolism
- Humans
- Lipoproteins, LDL/blood
- Low Density Lipoprotein Receptor-Related Protein-1
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microfilament Proteins/metabolism
- Muscle, Smooth, Vascular/cytology
- Mutation
- Myocytes, Smooth Muscle/metabolism
- Receptors, LDL/metabolism
- Receptors, Purinergic P2Y2/metabolism
- Signal Transduction
- Tumor Suppressor Proteins/metabolism
- Uridine Triphosphate/chemistry
Collapse
Affiliation(s)
| | - Cheikh I Seye
- Indiana University School of Medicine, Indianapolis, IN, United States
| | - Denis M Medeiros
- School of Graduate Studies, University of Missouri, Kansas City, MO, United States
| | - Gary A Weisman
- Department of Biochemistry and Bond Life Sciences Center, University of Missouri, Columbia, United States
| | - Barry Bradford
- Animal Sciences and Industry, Kansas State University, Manhattan, KS, United States
| | - Laman Mamedova
- Animal Sciences and Industry, Kansas State University, Manhattan, KS, United States.
| |
Collapse
|
16
|
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: 1.4] [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
|
17
|
Tidyman WE, Rauen KA. Pathogenetics of the RASopathies. Hum Mol Genet 2016; 25:R123-R132. [PMID: 27412009 DOI: 10.1093/hmg/ddw191] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 06/15/2016] [Indexed: 01/26/2023] Open
Abstract
The RASopathies are defined as a group of medical genetics syndromes that are caused by germ-line mutations in genes that encode components or regulators of the Ras/mitogen-activated protein kinase (MAPK) pathway. Taken together, the RASopathies represent one of the most prevalent groups of malformation syndromes affecting greater than 1 in 1,000 individuals. The Ras/MAPK pathway has been well studied in the context of cancer as it plays essential roles in growth, differentiation, cell cycle, senescence and apoptosis, all of which are also critical to normal development. The consequence of germ-line dysregulation leads to phenotypic alterations of development. RASopathies can be caused by several pathogenetic mechanisms that ultimately impact or alter the normal function and regulation of the MAPK pathway. These pathogenetic mechanisms can include functional alteration of GTPases, Ras GTPase-activating proteins, Ras guanine exchange factors, kinases, scaffolding or adaptor proteins, ubiquitin ligases, phosphatases and pathway inhibitors. Although these mechanisms are diverse, the common underlying biochemical phenotype shared by all the RASopathies is Ras/MAPK pathway activation. This results in the overlapping phenotypic features among these syndromes.
Collapse
Affiliation(s)
- William E Tidyman
- Division of Behavioral and Developmental Pediatrics, Department of Pediatrics UC Davis MIND Institute, Sacramento, CA 95817, USA
| | - Katherine A Rauen
- Department of Pediatrics, Division of Genomic Medicine, University of California Davis, Sacramento, CA, USA UC Davis MIND Institute, Sacramento, CA 95817, USA
| |
Collapse
|
18
|
Abstract
The Ras/mitogen activated protein kinase (MAPK) pathway is essential in the regulation of cell cycle, differentiation, growth, cell senescence and apoptosis, all of which are critical to normal development. A class of neurodevelopmental disorders, RASopathies, is caused by germline mutations in genes of the Ras/MAPK pathway. Through the use of whole exome sequencing and targeted sequencing of selected genes in cohorts of panel-negative RASopathy patients, several new genes have been identified. These include: RIT1, SOS2, RASA2, RRAS and SYNGAP1, that likely represent new, albeit rare, causative RASopathy genes. In addition, A2ML1, LZTR1, MYST4, SPRY1 and MAP3K8 may represent new rare genes for RASopathies, but, additional functional studies regarding the mutations are warranted. In addition, recent reports have demonstrated that chromosomal copy number variation in regions encompassing Ras/MAPK pathway genes may be a novel pathogenetic mechanism expanding the RASopathies.
Collapse
|
19
|
Ikoma-Seki K, Nakamura K, Morishita S, Ono T, Sugiyama K, Nishino H, Hirano H, Murakoshi M. Role of LRP1 and ERK and cAMP Signaling Pathways in Lactoferrin-Induced Lipolysis in Mature Rat Adipocytes. PLoS One 2015; 10:e0141378. [PMID: 26506094 PMCID: PMC4623961 DOI: 10.1371/journal.pone.0141378] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 10/06/2015] [Indexed: 01/14/2023] Open
Abstract
Lactoferrin (LF) is a multifunctional glycoprotein present in milk. A clinical study showed that enteric-coated bovine LF tablets decrease visceral fat accumulation. Furthermore, animal studies revealed that ingested LF is partially delivered to mesenteric fat, and in vitro studies showed that LF promotes lipolysis in mature adipocytes. The aim of the present study was to determine the mechanism underlying the induction of lipolysis in mature adipocytes that is induced by LF. To address this question, we used proteomics techniques to analyze protein expression profiles. Mature adipocytes from primary cultures of rat mesenteric fat were collected at various times after exposure to LF. Proteomic analysis revealed that the expression levels of hormone-sensitive lipase (HSL), which catalyzes the rate-limiting step of lipolysis, were upregulated and that HSL was activated by protein kinase A within 15 min after the cells were treated with LF. We previously reported that LF increases the intracellular concentration of cyclic adenosine monophosphate (cAMP), suggesting that LF activates the cAMP signaling pathway. In this study, we show that the expression level and the activity of the components of the extracellular signal-regulated kinase (ERK) signaling pathway were upregulated. Moreover, LF increased the activity of the transcription factor cAMP response element binding protein (CREB), which acts downstream in the cAMP and ERK signaling pathways and regulates the expression levels of adenylyl cyclase and HSL. Moreover, silencing of the putative LF receptor low-density lipoprotein receptor-related protein 1 (LRP1) attenuated lipolysis in LF-treated adipocytes. These results suggest that LF promoted lipolysis in mature adipocytes by regulating the expression levels of proteins involved in lipolysis through controlling the activity of cAMP/ERK signaling pathways via LRP1.
Collapse
Affiliation(s)
- Keiko Ikoma-Seki
- Research and Development Headquarters, Lion Corporation, Kanagawa, Japan
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
- * E-mail:
| | - Kanae Nakamura
- Research and Development Headquarters, Lion Corporation, Kanagawa, Japan
| | - Satoru Morishita
- Research and Development Headquarters, Lion Corporation, Kanagawa, Japan
| | - Tomoji Ono
- Research and Development Headquarters, Lion Corporation, Kanagawa, Japan
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Keikichi Sugiyama
- Research and Development Headquarters, Lion Corporation, Kanagawa, Japan
- Ritsumeikan University, Shiga, Japan
| | - Hoyoku Nishino
- Kyoto Prefectural University of Medicine, Kyoto, Japan
- Ritsumeikan University, Shiga, Japan
| | - Hisashi Hirano
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Michiaki Murakoshi
- Research and Development Headquarters, Lion Corporation, Kanagawa, Japan
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
- Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
20
|
Lin L, Hu K. LRP-1: functions, signaling and implications in kidney and other diseases. Int J Mol Sci 2014; 15:22887-901. [PMID: 25514242 PMCID: PMC4284744 DOI: 10.3390/ijms151222887] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 11/06/2014] [Accepted: 12/04/2014] [Indexed: 12/17/2022] Open
Abstract
Low-density lipoprotein (LDL)-related protein-1 (LRP-1) is a member of LDL receptor family that is implicated in lipoprotein metabolism and in the homeostasis of proteases and protease inhibitors. Expression of LRP-1 is ubiquitous. Up-regulation of LRP-1 has been reported in numerous human diseases. In addition to its function as a scavenger receptor for various ligands, LRP-1 has been shown to transduce multiple intracellular signal pathways including mitogen-activated protein kinase (MAPK), Akt, Rho, and the integrin signaling. LRP-1 signaling plays an important role in the regulation of diverse cellular process, such as cell proliferation, survival, motility, differentiation, and transdifferentiation, and thus participates in the pathogenesis of organ dysfunction and injury. In this review, we focus on the current understanding of LRP-1 signaling and its roles in the development and progression of kidney disease. The role and signaling of LRP-1 in the nervous and cardiovascular systems, as well as in carcinogenesis, are also briefly discussed.
Collapse
Affiliation(s)
- Ling Lin
- Division of Nephrology, Department of Medicine, College of Medicine, Penn State University, 500 University Drive, Hershey, PA 17033, USA.
| | - Kebin Hu
- Division of Nephrology, Department of Medicine, College of Medicine, Penn State University, 500 University Drive, Hershey, PA 17033, USA.
| |
Collapse
|
21
|
Güngör C, Hofmann BT, Wolters-Eisfeld G, Bockhorn M. Pancreatic cancer. Br J Pharmacol 2014; 171:849-58. [PMID: 24024905 DOI: 10.1111/bph.12401] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 08/16/2013] [Accepted: 09/03/2013] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED In recent years, it has become clear that the current standard therapeutic options for pancreatic cancer are not adequate and still do not meet the criteria to cure patients suffering from this lethal disease. Although research over the past decade has shown very interesting and promising new therapeutic options for these patients, only minor clinical success was achieved. Therefore, there is still an urgent need for new approaches that deal with early detection and new therapeutic options in pancreatic cancer. To provide optimal care for patients with pancreatic cancer, we need to understand better its complex molecular biology and thus to identify new target molecules that promote the proliferation and resistance to chemotherapy of pancreatic cancer cells. In spite of significant progress in curing cancers with chemotherapy, pancreatic cancer remains one of the most resistant solid tumour cancers and many studies suggest that drug-resistant cancer cells are the most aggressive with the highest relapse and metastatic rates. In this context, activated Notch signalling is strongly linked with chemoresistance and therefore reflects a rational new target to circumvent resistance to chemotherapy in pancreatic cancer. Here, we have focused our discussion on the latest research, current therapy options and recently identified target molecules such as Notch-2 and the heparin-binding growth factor midkine, which exhibit a wide range of cancer-relevant functions and therefore provide attractive new therapeutic target molecules, in terms of pancreatic cancer and other cancers also. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- C Güngör
- Department of General, Visceral and Thoracic Surgery, Experimental Oncology, Campus Research, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | |
Collapse
|
22
|
Abstract
Proteins generally act by binding to other molecules, including proteins. When proteins bind to other proteins, we speak of protein-protein interactions. It has become apparent that protein-protein interactions are critically important to many processes that take place in the cell, including signal transduction, regulation of gene expression, vesicular transport, nuclear import and export, and cell migration (Pawson and Nash, 2003). This has led to the recognition of protein-protein interactions as targets for drug development and to an increased interest in the identification of novel protein-protein interactions (Fry and Vassilev, 2005; Fry, 2006; Tord et al., 2007). Coimmunoprecipitation is a technique that is used to confirm novel protein-protein interactions in the context of a living cell or organism. In addition, coimmunoprecipitation is also used to study the dynamics of protein-protein interactions in response to intra- or extracellular stimuli, or can be used to study the effect of mutations on the ability of a protein to engage its binding partner. In a coimmunoprecipitation experiment, a protein of interest is isolated by immunoprecipitation. Subsequently, the presence of binding partners can be assessed by immunoblotting (see Western Blotting using Chemiluminescent Substrates).
Collapse
Affiliation(s)
- Peter van der Geer
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, CA, USA.
| |
Collapse
|
23
|
Beloglazova IB, Beabealashvilli RS, Gursky YG, Bocharov EV, Mineev KS, Parfenova EV, Tkachuk VA. Structural investigations of recombinant urokinase growth factor-like domain. BIOCHEMISTRY (MOSCOW) 2013; 78:517-30. [PMID: 23848154 DOI: 10.1134/s0006297913050106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Urokinase-type plasminogen activator (uPA) is a serine protease that converts the plasminogen zymogen into the enzymatically active plasmin. uPA is synthesized and secreted as the single-chain molecule (scuPA) composed of an N-terminal domain (GFD) and kringle (KD) and C-terminal proteolytic (PD) domains. Earlier, the structure of ATF (which consists of GFD and KD) was solved by NMR (A. P. Hansen et al. (1994) Biochemistry, 33, 4847-4864) and by X-ray crystallography alone and in a complex with the soluble form of the urokinase receptor (uPAR, CD87) lacking GPI (C. Barinka et al. (2006) J. Mol. Biol., 363, 482-495). According to these data, GFD contains two β-sheet regions oriented perpendicularly to each other. The area in the GFD responsible for binding to uPAR is localized in the flexible Ω-loop, which consists of seven amino acid residues connecting two strings of antiparallel β-sheet. It was shown by site-directed mutagenesis that shortening of the Ω-loop length by one amino acid residue leads to the inability of GFD to bind to uPAR (V. Magdolen et al. (1996) Eur. J. Biochem., 237, 743-751). Here we show that, in contrast to the above-mentioned studies, we found no sign of the β-sheet regions in GFD in our uPA preparations either free or in a complex with uPAR. The GFD seems to be a rather flexible and unstructured domain, demonstrating in spite of its apparent flexibility highly specific interaction with uPAR both in vitro and in cell culture experiments. Circular dichroism, tryptophan fluorescence during thermal denaturation of the protein, and heteronuclear NMR spectroscopy of ¹⁵N/¹³C-labeled ATF both free and in complex with urokinase receptor were used to judge the secondary structure of GFD of uPA.
Collapse
Affiliation(s)
- I B Beloglazova
- Russian Cardiology Research and Production Complex, 3-ya Cherepkovskaya ul. 15a, 121552 Moscow, Russia
| | | | | | | | | | | | | |
Collapse
|
24
|
Craig J, Mikhailenko I, Noyes N, Migliorini M, Strickland DK. The LDL receptor-related protein 1 (LRP1) regulates the PDGF signaling pathway by binding the protein phosphatase SHP-2 and modulating SHP-2- mediated PDGF signaling events. PLoS One 2013; 8:e70432. [PMID: 23922991 PMCID: PMC3724782 DOI: 10.1371/journal.pone.0070432] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 06/18/2013] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The PDGF signaling pathway plays a major role in several biological systems, including vascular remodeling that occurs following percutaneous transluminal coronary angioplasty. Recent studies have shown that the LDL receptor-related protein 1 (LRP1) is a physiological regulator of the PDGF signaling pathway. The underlying mechanistic details of how this regulation occurs have yet to be resolved. Activation of the PDGF receptor β (PDGFRβ) leads to tyrosine phosphorylation of the LRP1 cytoplasmic domain within endosomes and generates an LRP1 molecule with increased affinity for adaptor proteins such as SHP-2 that are involved in signaling pathways. SHP-2 is a protein tyrosine phosphatase that positively regulates the PDGFRβ pathway, and is required for PDGF-mediated chemotaxis. We investigated the possibility that LRP1 may regulate the PDGFRβ signaling pathway by binding SHP-2 and competing with the PDGFRβ for this molecule. METHODOLOGY/PRINCIPAL FINDINGS To quantify the interaction between SHP-2 and phosphorylated forms of the LRP1 intracellular domain, we utilized an ELISA with purified recombinant proteins. These studies revealed high affinity binding of SHP-2 to phosphorylated forms of both LRP1 intracellular domain and the PDGFRβ kinase domain. By employing the well characterized dynamin inhibitor, dynasore, we established that PDGF-induced SHP-2 phosphorylation primarily occurs within endosomal compartments, the same compartments in which LRP1 is tyrosine phosphorylated by activated PDGFRβ. Immunofluorescence studies revealed colocalization of LRP1 and phospho-SHP-2 following PDGF stimulation of fibroblasts. To define the contribution of LRP1 to SHP-2-mediated PDGF chemotaxis, we employed fibroblasts expressing LRP1 and deficient in LRP1 and a specific SHP-2 inhibitor, NSC-87877. Our results reveal that LRP1 modulates SHP-2-mediated PDGF-mediated chemotaxis. CONCLUSIONS/SIGNIFICANCE Our data demonstrate that phosphorylated forms of LRP1 and PDGFRβ compete for SHP-2 binding, and that expression of LRP1 attenuates SHP-2-mediated PDGF signaling events.
Collapse
Affiliation(s)
- Julie Craig
- Center for Vascular and Inflammatory Diseases and
| | - Irina Mikhailenko
- Center for Vascular and Inflammatory Diseases and
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | | | - Mary Migliorini
- Center for Vascular and Inflammatory Diseases and
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Dudley K. Strickland
- Center for Vascular and Inflammatory Diseases and
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
25
|
Stewart CE, Sayers I. Urokinase receptor orchestrates the plasminogen system in airway epithelial cell function. Lung 2013; 191:215-25. [PMID: 23408042 DOI: 10.1007/s00408-013-9450-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 01/10/2013] [Indexed: 11/24/2022]
Abstract
PURPOSE The plasminogen system plays many roles in normal epithelial cell function, and components are elevated in diseases, such as cancer and asthma. The relative contribution of each component to epithelial function is unclear. We characterized normal cell function in airway epithelial cells with increased expression of selected pathway components. METHODS BEAS-2B R1 bronchial epithelial cells stably overexpressing membrane urokinase plasminogen activator receptor (muPAR), soluble spliced uPAR (ssuPAR), the ligand (uPA) or inhibitors (PAI1 or PAI2), were characterized for pathway expression. Cell function was examined using proliferation, apoptosis, and scratch wound assays. A549 alveolar epithelial cells overexpressing muPAR were similarly characterized and downstream plasmin activity, MMP-1, and MMP-9 measured. RESULTS Elevated expression of individual components led to changes in the plasminogen system expression profile, indicating coordinated regulation of the pathway. Increased muPAR expression augmented wound healing rate in BEAS-2B R1 and attenuated repair in A549 cells. Elevated expression of other system components had no effect on cell function in BEAS-2B R1 cells. This is the first study to investigate activity of the splice variant ssuPAR, with results suggesting that this variant plays a limited role in epithelial cell function in this model. CONCLUSIONS Our data highlight muPAR as the critical molecule orchestrating effects of the plasminogen system on airway epithelial cell function. These data have implications for diseases, such as cancer and asthma, and suggest uPAR as the key therapeutic target for the pathway in approaches to alter epithelial cell function.
Collapse
Affiliation(s)
- Ceri E Stewart
- Division of Therapeutics and Molecular Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | | |
Collapse
|
26
|
Sagare AP, Deane R, Zlokovic BV. Low-density lipoprotein receptor-related protein 1: a physiological Aβ homeostatic mechanism with multiple therapeutic opportunities. Pharmacol Ther 2012; 136:94-105. [PMID: 22820095 PMCID: PMC3432694 DOI: 10.1016/j.pharmthera.2012.07.008] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 07/03/2012] [Indexed: 11/29/2022]
Abstract
Low-density lipoprotein receptor-related protein-1 (LRP1) is the main cell surface receptor involved in brain and systemic clearance of the Alzheimer's disease (AD) toxin amyloid-beta (Aβ). In plasma, a soluble form of LRP1 (sLRP1) is the major transport protein for peripheral Aβ. LRP1 in brain endothelium and mural cells mediates Aβ efflux from brain by providing a transport mechanism for Aβ across the blood-brain barrier (BBB). sLRP1 maintains a plasma 'sink' activity for Aβ through binding of peripheral Aβ which in turn inhibits re-entry of free plasma Aβ into the brain. LRP1 in the liver mediates systemic clearance of Aβ. In AD, LRP1 expression at the BBB is reduced and Aβ binding to circulating sLRP1 is compromised by oxidation. Cell surface LRP1 and circulating sLRP1 represent druggable targets which can be therapeutically modified to restore the physiological mechanisms of brain Aβ homeostasis. In this review, we discuss how increasing LRP1 expression at the BBB and liver with lifestyle changes, statins, plant-based active principles and/or gene therapy on one hand, and how replacing dysfunctional plasma sLRP1 on the other regulate Aβ clearance from brain ultimately controlling the onset and/or progression of AD.
Collapse
Affiliation(s)
- Abhay P. Sagare
- Department of Physiology and Biophysics, and Center for Neurodegeneration and Regeneration at the Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, 1501 San Pablo Street, Los Angeles, CA 90089, United States
| | - Rashid Deane
- Department of Neurosurgery, Arthur Kornberg Medical Research Building, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Berislav V. Zlokovic
- Department of Physiology and Biophysics, and Center for Neurodegeneration and Regeneration at the Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, 1501 San Pablo Street, Los Angeles, CA 90089, United States
| |
Collapse
|
27
|
Eastman BM, Jo M, Webb DL, Takimoto S, Gonias SL. A transformation in the mechanism by which the urokinase receptor signals provides a selection advantage for estrogen receptor-expressing breast cancer cells in the absence of estrogen. Cell Signal 2012; 24:1847-55. [PMID: 22617030 DOI: 10.1016/j.cellsig.2012.05.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 05/08/2012] [Accepted: 05/13/2012] [Indexed: 10/28/2022]
Abstract
Binding of urokinase-type plasminogen activator (uPA) to its receptor, uPAR, in estrogen receptor-α (ERα) expressing breast cancer cells, transiently activates ERK downstream of FAK, Src family kinases, and H-Ras. Herein, we show that when uPAR is over-expressed, in two separate ERα-positive breast cancer cell lines, ERK activation occurs autonomously of uPA and is sustained. Autonomous ERK activation by uPAR requires H-Ras and Rac1. A mutated form of uPAR, which does not bind vitronectin (uPAR-W32A), failed to induce autonomous ERK activation. Expression of human uPAR or mouse uPAR but not uPAR-W32A in MCF-7 cells provided a selection advantage when these cells were deprived of estrogen in cell culture for two weeks. Similarly, MCF-7 cells that express mouse uPAR formed xenografts in SCID mice that survived and increased in volume in the absence of estrogen supplementation, probably reflecting the pro-survival activity of phospho-ERK. Autonomous uPAR signaling to ERK was sensitive to the EGFR tyrosine kinase inhibitors, Erlotinib and Gefitinib. The transition in uPAR signaling from uPA-dependent and transient to autonomous and sustained is reminiscent of the transformation in ErbB2/HER2 signaling observed when this gene is amplified in breast cancer. uPAR over-expression may provide a pathway for escape of breast cancer cells from ERα-targeting therapeutics.
Collapse
Affiliation(s)
- Boryana M Eastman
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA 92093‐0612, USA
| | | | | | | | | |
Collapse
|
28
|
Novel aspects of the apolipoprotein-E receptor family: regulation and functional role of their proteolytic processing. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/s11515-011-1186-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
29
|
Huang XY, Shi GM, Devbhandari RP, Ke AW, Wang Y, Wang XY, Wang Z, Shi YH, Xiao YS, Ding ZB, Dai Z, Xu Y, Jia WP, Tang ZY, Fan J, Zhou J. Low level of low-density lipoprotein receptor-related protein 1 predicts an unfavorable prognosis of hepatocellular carcinoma after curative resection. PLoS One 2012; 7:e32775. [PMID: 22427881 PMCID: PMC3299691 DOI: 10.1371/journal.pone.0032775] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Accepted: 01/30/2012] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional receptor involved in receptor-mediated endocytosis and cell signaling. The aim of this study was to elucidate the expression and mechanism of LRP1 in hepatocellular carcinoma (HCC). METHODS LRP1 expression in 4 HCC cell lines and 40 HCC samples was detected. After interruption of LRP1 expression in a HCC cell line either with specific lentiviral-mediated shRNA LRP1 or in the presence of the LRP1-specific chaperone, receptor-associated protein (RAP), the role of LRP1 in the migration and invasion of HCC cells was assessed in vivo and in vitro, and the expression of matrix metalloproteinase (MMP) 9 in cells and the bioactivity of MMP9 in the supernatant were assayed. The expression and prognostic value of LRP1 were investigated in 327 HCC specimens. RESULTS Low LRP1 expression was associated with poor HCC prognosis, with low expression independently related to shortened overall survival and increased tumor recurrence rate. Expression of LRP1 in non-recurrent HCC samples was significantly higher than that in early recurrent samples. LRP1 expression in HCC cell lines was inversely correlated with their metastatic potential. After inhibition of LRP1, low-metastatic SMCC-7721 cells showed enhanced migration and invasion and increased expression and bioactivity of MMP9. Correlation analysis showed a negative correlation between LRP1 and MMP9 expression in HCC patients. The prognostic value of LRP1 expression was validated in the independent data set. CONCLUSIONS LRP1 modulated the level of MMP9 and low level of LRP1 expression was associated with aggressiveness and invasiveness in HCCs. LRP1 offered a possible strategy for tumor molecular therapy.
Collapse
Affiliation(s)
- Xiao-Yong Huang
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Organ Transplantation, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai, People's Republic of China,
| | - Guo-Ming Shi
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Organ Transplantation, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | | | - Ai-Wu Ke
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Yuwei Wang
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Xiao-Ying Wang
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Zheng Wang
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Ying-Hong Shi
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Yong-Sheng Xiao
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Zhen-Bin Ding
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Zhi Dai
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Organ Transplantation, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Yang Xu
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Wei-Ping Jia
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University, Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Zhao-You Tang
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
| | - Jia Fan
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Organ Transplantation, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Cancer Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai, People's Republic of China,
- * E-mail: (JF); (JZ)
| | - Jian Zhou
- Liver Cancer Institute, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Organ Transplantation, Fudan University, Zhongshan Hospital, Shanghai, People's Republic of China
- Cancer Center, Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai, People's Republic of China,
- * E-mail: (JF); (JZ)
| |
Collapse
|
30
|
Cathepsin D is partly endocytosed by the LRP1 receptor and inhibits LRP1-regulated intramembrane proteolysis. Oncogene 2011; 31:3202-12. [PMID: 22081071 DOI: 10.1038/onc.2011.501] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The aspartic protease cathepsin-D (cath-D) is a marker of poor prognosis in breast cancer that is overexpressed and hypersecreted by human breast cancer cells. Secreted pro-cath-D binds to the extracellular domain of the β-chain of the LDL receptor-related protein-1 (LRP1) in fibroblasts. The LRP1 receptor has an 85-kDa transmembrane β-chain and a noncovalently attached 515-kDa extracellular α-chain. LRP1 acts by (1) internalizing many ligands via its α-chain, (2) activating signaling pathways by phosphorylating the LRP1β-chain tyrosine and (3) modulating gene transcription by regulated intramembrane proteolysis (RIP) of its β-chain. LRP1 RIP involves two cleavages: the first liberates the LRP1 ectodomain to give a membrane-associated form, LRP1β-CTF, and the second generates the LRP1β-intracellular domain, LRP1β-ICD, that modulates gene transcription. Here, we investigated the endocytosis of pro-cath-D by LRP1 and the effect of pro-cath-D/LRP1β interaction on LRP1β tyrosine phosphorylation and/or LRP1β RIP. Our results indicate that pro-cath-D was partially endocytosed by LRP1 in fibroblasts. However, pro-cath-D and ectopic cath-D did not stimulate phosphorylation of the LRP1β-chain tyrosine. Interestingly, ectopic cath-D and its catalytically inactive (D231N)cath-D, and pro-(D231N)cath-D all significantly inhibited LRP1 RIP by preventing LRP1β-CTF production. Thus, cath-D inhibits LRP1 RIP independently of its catalytic activity by blocking the first cleavage. As cath-D triggers fibroblast outgrowth by LRP1, we propose that cath-D modulates the growth of fibroblasts by inhibiting LRP1 RIP in the breast tumor microenvironment.
Collapse
|
31
|
Pawaria S, Binder RJ. CD91-dependent programming of T-helper cell responses following heat shock protein immunization. Nat Commun 2011; 2:521. [PMID: 22045000 DOI: 10.1038/ncomms1524] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 10/03/2011] [Indexed: 12/18/2022] Open
Abstract
The immunogenic heat shock proteins (HSPs) gp96, hsp70 and calreticulin (CRT) bind to CD91 on antigen-presenting cells (APCs) for cross-presentation of the HSP-chaperoned peptides. This event leads to priming of T-cell responses. Here we show that CD91 serves as a signalling receptor for these HSPs, allowing for the maturation of APCs, secretion of cytokines and priming of T-helper (Th) cells. Specifically, CD91 is phosphorylated in response to HSPs in a unique pattern and phospho-CD91 triggers signalling cascades to activate nuclear factor-kappa B. Each HSP-CD91 interaction on APCs stimulates a unique cytokine profile, which dictates priming of specific Th cell subsets. Thus, in a transforming growth factor-β tumour microenvironment, immunization with CRT, but not gp96 or hsp70, primes Th17-cell responses in a CD91-dependent manner. These results are important for development of T-cell responses in situ in tumour-bearing hosts and for vaccination against cancer and infectious disease.
Collapse
Affiliation(s)
- Sudesh Pawaria
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | | |
Collapse
|
32
|
Dieckmann M, Dietrich MF, Herz J. Lipoprotein receptors--an evolutionarily ancient multifunctional receptor family. Biol Chem 2011; 391:1341-63. [PMID: 20868222 DOI: 10.1515/bc.2010.129] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The evolutionarily ancient low-density lipoprotein (LDL) receptor gene family represents a class of widely expressed cell surface receptors. Since the dawn of the first primitive multicellular organisms, several structurally and functionally distinct families of lipoprotein receptors have evolved. In accordance with the now obsolete 'one-gene-one-function' hypothesis, these cell surface receptors were originally perceived as mere transporters of lipoproteins, lipids, and nutrients or as scavenger receptors, which remove other kinds of macromolecules, such as proteases and protease inhibitors from the extracellular environment and the cell surface. This picture has since undergone a fundamental change. Experimental evidence has replaced the perception that these receptors serve merely as cargo transporters. Instead it is now clear that the transport of macromolecules is inseparably intertwined with the molecular machinery by which cells communicate with each other. Lipoprotein receptors are essentially sensors of the extracellular environment that participate in a wide range of physiological processes by physically interacting and coevolving with primary signal transducers as co-regulators. Furthermore, lipoprotein receptors modulate cellular trafficking and localization of the amyloid precursor protein (APP) and the β-amyloid peptide (Aβ), suggesting a role in the pathogenesis of Alzheimer's disease. Moreover, compelling evidence shows that LDL receptor family members are involved in tumor development and progression.
Collapse
Affiliation(s)
- Marco Dieckmann
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9046, USA
| | | | | |
Collapse
|
33
|
Boucher P, Herz J. Signaling through LRP1: Protection from atherosclerosis and beyond. Biochem Pharmacol 2010; 81:1-5. [PMID: 20920479 DOI: 10.1016/j.bcp.2010.09.018] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 09/14/2010] [Accepted: 09/20/2010] [Indexed: 11/18/2022]
Abstract
The low-density lipoprotein receptor-related protein (LRP1) is a multifunctional cell surface receptor that belongs to the LDL receptor (LDLR) gene family and that is widely expressed in several tissues. LRP1 consists of an 85-kDa membrane-bound carboxyl fragment (β chain) and a non-covalently attached 515-kDa (α chain) amino-terminal fragment. Through its extracellular domain, LRP1 binds at least 40 different ligands ranging from lipoprotein and protease inhibitor complex to growth factors and extracellular matrix proteins. LRP-1 has also been shown to interact with scaffolding and signaling proteins via its intracellular domain in a phosphorylation-dependent manner and to function as a co-receptor partnering with other cell surface or integral membrane proteins. LRP-1 is thus implicated in two major physiological processes: endocytosis and regulation of signaling pathways, which are both involved in diverse biological roles including lipid metabolism, cell growth/differentiation processes, degradation of proteases, and tissue invasion. The embryonic lethal phenotype obtained after target disruption of the LRP-1 gene in the mouse highlights the biological importance of this receptor and revealed a critical, but yet undefined role in development. Tissue-specific gene deletion studies also reveal an important contribution of LRP1 in vascular remodeling, foam cell biology, the central nervous system, and in the molecular mechanisms of atherosclerosis.
Collapse
Affiliation(s)
- Philippe Boucher
- CNRS, UMR7175, Université de Strasbourg, 74, route du Rhin, Illkirch F-67401, France.
| | | |
Collapse
|
34
|
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: 1.4] [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
|
35
|
Lin L, Bu G, Mars WM, Reeves WB, Tanaka S, Hu K. tPA activates LDL receptor-related protein 1-mediated mitogenic signaling involving the p90RSK and GSK3beta pathway. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:1687-96. [PMID: 20724593 DOI: 10.2353/ajpath.2010.100213] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In renal fibrosis, interstitial fibroblasts have an increased proliferative phenotype, and the numbers of interstitial fibroblasts closely correlate with the extent of kidney damage. The mechanisms underlying proliferation and resulting expansion of the interstitium remain largely unknown. Here we define the intracellular signaling events by which tissue plasminogen activator (tPA) promotes renal interstitial fibroblast proliferation. tPA promoted the proliferation of renal interstitial fibroblasts independent of its protease activity. The mitogenic effect of tPA required Tyr(4507) phosphorylation of the cytoplasmic tail of its receptor LDL receptor-related protein 1. tPA triggered sequential proliferative signaling events involving Erk1/2, p90RSK, GSK3β phosphorylation, and cyclin D1 induction. Blockade of Erk1/2 activation or knockdown of p90RSK suppressed tPA-induced GSK3β phosphorylation, cyclin D1 expression, and fibroblast proliferation. In contrast, expression of constitutively active Mek1 mimicked tPA in inducing GSK3β phosphorylation and cyclin D1 expression. Ectopic overexpression of an uninhibitable GSK3β mutant eliminated tPA-induced cyclin D1 expression. In the murine obstruction model, tPA deficiency reduced renal GSK3β phosphorylation and induction of PCNA and FSP-1. These findings show that tPA induces Tyr(4507) phosphorylation of LDL receptor-related protein 1, which in turn leads to the downstream phosphorylation of Erk1/2, p90RSK, and GSK3β, followed by the induction of cyclin D1 in murine interstitial fibroblasts. This study implicates tPA as a mitogen that promotes interstitial fibroblast proliferation, leading to expansion of these cells.
Collapse
Affiliation(s)
- Ling Lin
- Division of Nephrology, Department of Medicine, Penn State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | | | | | | | | | | |
Collapse
|
36
|
Langlois B, Perrot G, Schneider C, Henriet P, Emonard H, Martiny L, Dedieu S. LRP-1 promotes cancer cell invasion by supporting ERK and inhibiting JNK signaling pathways. PLoS One 2010; 5:e11584. [PMID: 20644732 PMCID: PMC2904376 DOI: 10.1371/journal.pone.0011584] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Accepted: 06/20/2010] [Indexed: 01/16/2023] Open
Abstract
Background The low-density lipoprotein receptor-related protein-1 (LRP-1) is an endocytic receptor mediating the clearance of various extracellular molecules involved in the dissemination of cancer cells. LRP-1 thus appeared as an attractive receptor for targeting the invasive behavior of malignant cells. However, recent results suggest that LRP-1 may facilitate the development and growth of cancer metastases in vivo, but the precise contribution of the receptor during cancer progression remains to be elucidated. The lack of mechanistic insights into the intracellular signaling networks downstream of LRP-1 has prevented the understanding of its contribution towards cancer. Methodology/Principal Findings Through a short-hairpin RNA-mediated silencing approach, we identified LRP-1 as a main regulator of ERK and JNK signaling in a tumor cell context. Co-immunoprecipitation experiments revealed that LRP-1 constitutes an intracellular docking site for MAPK containing complexes. By using pharmacological agents, constitutively active and dominant-negative kinases, we demonstrated that LRP-1 maintains malignant cells in an adhesive state that is favorable for invasion by activating ERK and inhibiting JNK. We further demonstrated that the LRP-1-dependent regulation of MAPK signaling organizes the cytoskeletal architecture and mediates adhesive complex turnover in cancer cells. Moreover, we found that LRP-1 is tethered to the actin network and to focal adhesion sites and controls ERK and JNK targeting to talin-rich structures. Conclusions We identified ERK and JNK as the main molecular relays by which LRP-1 regulates focal adhesion disassembly of malignant cells to support invasion.
Collapse
Affiliation(s)
- Benoit Langlois
- Université de Reims Champagne-Ardenne, CNRS UMR 6237 MEDyC, Laboratoire SiRMa, Campus Moulin de la Housse, Reims, France
| | - Gwenn Perrot
- Université de Reims Champagne-Ardenne, CNRS UMR 6237 MEDyC, Laboratoire SiRMa, Campus Moulin de la Housse, Reims, France
| | - Christophe Schneider
- Université de Reims Champagne-Ardenne, CNRS UMR 6237 MEDyC, Laboratoire SiRMa, Campus Moulin de la Housse, Reims, France
| | - Patrick Henriet
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium
| | - Hervé Emonard
- Université de Reims Champagne-Ardenne, CNRS UMR 6237 MEDyC, Laboratoire SiRMa, Campus Moulin de la Housse, Reims, France
| | - Laurent Martiny
- Université de Reims Champagne-Ardenne, CNRS UMR 6237 MEDyC, Laboratoire SiRMa, Campus Moulin de la Housse, Reims, France
| | - Stéphane Dedieu
- Université de Reims Champagne-Ardenne, CNRS UMR 6237 MEDyC, Laboratoire SiRMa, Campus Moulin de la Housse, Reims, France
- * E-mail:
| |
Collapse
|
37
|
Jeong YH, Ishikawa K, Someya Y, Hosoda A, Yoshimi T, Yokoyama C, Kiryu-Seo S, Kang MJ, Tchibana T, Kiyama H, Fukumura T, Kim DH, Saeki S. Molecular characterization and expression of the low-density lipoprotein receptor-related protein-10, a new member of the LDLR gene family. Biochem Biophys Res Commun 2010; 391:1110-5. [DOI: 10.1016/j.bbrc.2009.12.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Accepted: 12/08/2009] [Indexed: 10/20/2022]
|
38
|
Reekmans SM, Pflanzner T, Gordts PLSM, Isbert S, Zimmermann P, Annaert W, Weggen S, Roebroek AJM, Pietrzik CU. Inactivation of the proximal NPXY motif impairs early steps in LRP1 biosynthesis. Cell Mol Life Sci 2010; 67:135-45. [PMID: 19856143 PMCID: PMC11115674 DOI: 10.1007/s00018-009-0171-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 09/24/2009] [Accepted: 10/05/2009] [Indexed: 11/25/2022]
Abstract
The proximal NPXY and distal NPXYXXL motifs in the intracellular domain of LRP1 play an important role in regulation of the function of the receptor. The impact of single and double inactivating knock-in mutations of these motifs on receptor maturation, cell surface expression, and ligand internalization was analyzed in mutant and control wild-type mice and MEFs. Single inactivation of the proximal NPXY or in combination with inactivation of the distal NPXYXXL motif are both shown to be associated with an impaired maturation and premature proteasomal degradation of full-length LRP1. Therefore, only a small mature LRP1 pool is able to reach the cell surface resulting indirectly in severe impairment of ligand internalization. Single inactivation of the NPXYXXL motif revealed normal maturation, but direct impairment of ligand internalization. In conclusion, the proximal NPXY motif proves to be essential for early steps in the LRP1 biosynthesis, whereas NPXYXXL appears rather relevant for internalization.
Collapse
Affiliation(s)
- Sara M. Reekmans
- Laboratory for Experimental Mouse Genetics, Center for Human Genetics, KU Leuven, Herestraat 49, bus 602, 3000 Leuven, Belgium
- Laboratory for Experimental Mouse Genetics, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium
| | - Thorsten Pflanzner
- Molecular Neurodegeneration, Department of Physiological Chemistry and Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Duesbergweg 6, 55099 Mainz, Germany
| | - Philip L. S. M. Gordts
- Laboratory for Experimental Mouse Genetics, Center for Human Genetics, KU Leuven, Herestraat 49, bus 602, 3000 Leuven, Belgium
- Laboratory for Experimental Mouse Genetics, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium
| | - Simone Isbert
- Molecular Neurodegeneration, Department of Physiological Chemistry and Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Duesbergweg 6, 55099 Mainz, Germany
| | - Pascale Zimmermann
- Laboratory for Signal Integration in Cell Fate Decision, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Wim Annaert
- Laboratory of Membrane Trafficking, Center for Human Genetics, KU Leuven, Leuven, Belgium
- Laboratory of Membrane Trafficking, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium
| | - Sascha Weggen
- Department of Neuropathology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Anton J. M. Roebroek
- Laboratory for Experimental Mouse Genetics, Center for Human Genetics, KU Leuven, Herestraat 49, bus 602, 3000 Leuven, Belgium
- Laboratory for Experimental Mouse Genetics, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium
| | - Claus U. Pietrzik
- Molecular Neurodegeneration, Department of Physiological Chemistry and Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Duesbergweg 6, 55099 Mainz, Germany
| |
Collapse
|
39
|
Extravasale Proteolyse: Funktion und Interaktion der Faktoren des fibrinolytischen Systems. Hamostaseologie 2010. [DOI: 10.1007/978-3-642-01544-1_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
40
|
Guttman M, Betts GN, Barnes H, Ghassemian M, van der Geer P, Komives EA. Interactions of the NPXY microdomains of the low density lipoprotein receptor-related protein 1. Proteomics 2009; 9:5016-28. [PMID: 19771558 PMCID: PMC2862490 DOI: 10.1002/pmic.200900457] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The low density lipoprotein receptor-related protein 1 (LRP1) mediates internalization of a large number of proteins and protein-lipid complexes and is widely implicated in Alzheimer's disease. The cytoplasmic domain of LRP1 (LRP1-CT) can be phosphorylated by activated protein-tyrosine kinases at two NPXY motifs in LRP1-CT; Tyr 4507 is readily phosphorylated and must be phosphorylated before phosphorylation of Tyr 4473 occurs. Pull-down experiments from brain lysate revealed numerous proteins binding to LRP1-CT, but the results were highly variable. To separate which proteins bind to each NPXY motif and their phosphorylation dependence, each NPXY motif microdomain was prepared in both phosphorylated and non-phosphorylated forms and used to probe rodent brain extracts for binding proteins. Proteins that bound specifically to the microdomains were identified by LC-MS/MS, and confirmed by Western blot. Recombinant proteins were then tested for binding to each NPXY motif. The NPXY(4507) (membrane distal) was found to interact with a large number of proteins, many of which only bound the tyrosine-phosphorylated form. This microdomain also bound a significant number of other proteins in the unphosphorylated state. Many of the interactions were later confirmed to be direct with recombinant proteins. The NPXY(4473) (membrane proximal) bound many fewer proteins and only to the phosphorylated form.
Collapse
Affiliation(s)
- Miklos Guttman
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Dr. La Jolla, CA 92093-0378
| | - Gina N. Betts
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Dr. La Jolla, CA 92093-0378
| | - Helen Barnes
- Department of Chemistry and Biochemistry, San Diego State University, 5500 Campanile Dr. San Diego CA 92182-1030
| | | | - Peter van der Geer
- Department of Chemistry and Biochemistry, San Diego State University, 5500 Campanile Dr. San Diego CA 92182-1030
| | - Elizabeth A. Komives
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Dr. La Jolla, CA 92093-0378
| |
Collapse
|
41
|
Basford JE, Moore ZWQ, Zhou L, Herz J, Hui DY. Smooth muscle LDL receptor-related protein-1 inactivation reduces vascular reactivity and promotes injury-induced neointima formation. Arterioscler Thromb Vasc Biol 2009; 29:1772-8. [PMID: 19729608 DOI: 10.1161/atvbaha.109.194357] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Defective smooth muscle expression of LDL receptor-related protein-1 (Lrp1) increases atherosclerosis in hypercholesterolemic mice. This study explored the importance of smooth muscle Lrp1 expression under normolipidemic conditions. METHODS AND RESULTS Smooth muscle cells isolated from control (smLrp1(+/+)) and smooth muscle-specific Lrp1 knockout (smLrp1(-/-)) mice were characterized based on morphology, smooth muscle marker protein expression levels, and growth rates in vitro. Vascular functions were assessed by aortic constrictive response to agonist stimulation in situ and neointimal hyperplasia to carotid arterial injury in vivo. The smLrp1(-/-) smooth muscle cells displayed reduced alpha-actin and calponin expression and an accelerated growth rate attribtuable to sustained phosphorylation of platelet-derived growth factor receptor (PRGFR) and protein kinase B/Akt. Vasoconstrictive response to agonist stimulation was impaired in aortic rings isolated from smLrp1(-/-) mice. Injury-induced neointimal hyperplasia was significantly increased in smLrp1(-/-) mice. The increase in neointima was associated with corresponding elevated activation of PDGFR signaling pathway. CONCLUSIONS Smooth muscle expression of Lrp1 is important in maintaining normal vascular functions under normolipidemic conditions. The absence of Lrp1 expression results in greater smooth muscle cell proliferation, deficient contractile protein expression, impairment of vascular contractility, and promotion of denudation-induced neointimal hyperplasia.
Collapse
Affiliation(s)
- Joshua E Basford
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA
| | | | | | | | | |
Collapse
|
42
|
Gordts PLSM, Reekmans S, Lauwers A, Van Dongen A, Verbeek L, Roebroek AJM. Inactivation of the LRP1 intracellular NPxYxxL motif in LDLR-deficient mice enhances postprandial dyslipidemia and atherosclerosis. Arterioscler Thromb Vasc Biol 2009; 29:1258-64. [PMID: 19667105 DOI: 10.1161/atvbaha.109.192211] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The purpose of this study was to determine the significance of the intracellular NPxYxxL motif of LRP1 for the atheroprotective role of this multifunctional receptor. METHODS AND RESULTS LRP1 knock-in mice carrying an inactivating mutation in the NPxYxxL motif were crossed with LDLR-deficient mice, a model for atherosclerosis. In this LDLR(-/-) background the mutated mice showed a more atherogenic lipoprotein profile, which was associated with a decreased clearance of postprandial lipids because of a compromised endocytosis rate and reduced lipase activity. On an atherogenic diet LRP1 mutant mice revealed a 50% increased development of atherosclerosis. This aggravation was accompanied by an increase in smooth muscle cell (SMC) and collagen content and apoptotic cells in the lesions. The mutation showed, however, a limited impact on basal PDGFR-beta expression and signaling and the antimigratory property of apoE on PDGF-BB-stimulated SMCs. Additionally, levels of LRP1 atherogenic ligands, like MMP2, t-PA, FVIII, and the inflammatory ligand TNF-alpha showed to be significantly elevated. CONCLUSIONS These findings demonstrate that the NPxYxxL motif is essential for the atheroprotective role of LRP1. This motif is relevant for normal control of lipid metabolism and of atherogenic and inflammatory ligands, but has no pronounced effect on regulating PDGF-BB/PDGFR-beta signaling in SMCs.
Collapse
|
43
|
Ding S, Bond AE, Lemière F, Tuytten R, Esmans EL, Brenton AG, Dudley E, Newton RP. Online immobilized metal affinity chromatography/mass spectrometric analysis of changes elicited by cCMP in the murine brain phosphoproteome. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2008; 22:4129-4138. [PMID: 19023864 DOI: 10.1002/rcm.3834] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
An automated online immobilized metal affinity chromatography/high-performance liquid chromatography mass spectrometric (IMAC-HPLC/MS/MS) method was developed to study cytidine 3',5'-cyclic monophosphate (cCMP)-specific protein phosphorylation, analogous to a previously successful offline IMAC method using microvolume IMAC pipette tips. The optimized method identified murine brain phosphoproteins selectively modified by challenge with cCMP, using manual interpretation of the results to confirm both phosphorylation and selectivity of response to cCMP. A number of proteins identified by this strategy have potential roles in hyperproliferation, a previously reported response to elevated levels of cCMP.
Collapse
Affiliation(s)
- S Ding
- Department of Environmental and Molecular Biosciences, Swansea University, SOTEAS, Singleton Park, Swansea SA2 8PP, UK
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Makarova A, Bercury KK, Adams KW, Joyner D, Deng M, Spoelgen R, Koker M, Strickland DK, Hyman BT. The LDL receptor-related protein can form homo-dimers in neuronal cells. Neurosci Lett 2008; 442:91-5. [PMID: 18602448 DOI: 10.1016/j.neulet.2008.06.070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Revised: 06/13/2008] [Accepted: 06/18/2008] [Indexed: 10/21/2022]
Abstract
The ability of the low density lipoprotein receptor-related protein (LRP) to form homo-dimers was studied in mouse neuroblastoma and human neuroglioma cells as well as in primary cortical cultures from adult mouse brain. Homo-dimerization of LRP light chain (LC) was shown by several methods including co-immunoprecipitation, fluorescence lifetime imaging microscopy, and bimolecular fluorescence complementation assay. The requirement of intact NPXY motifs of LRP LC for homo-dimerization was ruled out by co-immunoprecipitation assay.
Collapse
Affiliation(s)
- Alexandra Makarova
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Pancreatic reg I binds MKP-1 and regulates cyclin D in pancreatic-derived cells. J Surg Res 2008; 150:137-43. [PMID: 18929742 DOI: 10.1016/j.jss.2008.03.047] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2008] [Revised: 03/07/2008] [Accepted: 03/28/2008] [Indexed: 11/21/2022]
Abstract
BACKGROUND The pancreatic regenerating (reg I) gene and its protein product are derived from acinar cells and are mitogenic to beta- and ductal cells. We studied the mechanism of this mitogenic response. MATERIALS AND METHODS ARIP (rat ductal) and RIN 1046-38 (rat beta-) cell lines were exposed to exogenous reg I in culture or transfected with a reg I expression vector. Mitogenesis was assessed by MTS assay (CellTiter 96; Promega, Inc., Madison, WI), and cellular mRNA was subjected to gene microarray analysis to determine signal transduction pathways. Yeast two-hybrid technology was then used to determine intracellular binding of reg I protein. RESULTS Cells exposed to exogenous reg I showed a mitogenic response; cells transfected with reg I expression vector showed inhibited growth. Microarray analysis of the former showed induction of cyclin pathways and mitogen-activated protein kinase phosphatase (MKP-1); cyclins were inhibited in the latter. Northern analysis confirmed gene induction of cyclin D1 and MKP-1; JNK was phosphorylated prior to expression of both. Yeast two-hybrid analysis confirmed a protein-protein interaction with MKP-1; this was confirmed by immunoprecipitation. CONCLUSIONS Pancreatic-derived cells exposed to reg I grow by activation of signal transduction pathways involving the mitogen-activated protein kinase phosphatases and cyclins, with concomitant induction of MKP-1. However, high intracellular levels of reg I lead to decreased growth, likely via a binding to and inactivation of MKP-1. Inhibition of cell growth, and possible induction of apoptosis, may lead to differentiation of these cells to other cell types.
Collapse
|
46
|
Betts GN, van der Geer P, Komives EA. Structural and functional consequences of tyrosine phosphorylation in the LRP1 cytoplasmic domain. J Biol Chem 2008; 283:15656-64. [PMID: 18381291 DOI: 10.1074/jbc.m709514200] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cytoplasmic domain of LRP1 contains two NPXY motifs that have been shown to interact with signaling proteins. In previous work, we showed that Tyr(4507) in the distal NPXY motif is phosphorylated by v-Src, whereas denaturation of the protein was required for phosphorylation of Tyr(4473) in the membraneproximal NPXY motif. Amide H/D exchange studies reveal that the distal NPXY motif is fully solvent-exposed, whereas the proximal one is not. Phosphopeptide mapping combined with in vitro and in vivo kinase experiments show that Tyr(4473) can be phosphorylated, but only if Tyr(4507) is phosphorylated or substituted with glutamic acid. Amide H/D exchange experiments indicate that solvent accessibility increases across the entire LRP1 cytoplasmic region upon phosphorylation at Tyr(4507); in particular the NPXY(4473) motif becomes much more exposed. This differential phosphorylation is functionally relevant: binding of Snx17, which is known to bind at the proximal NPXY motif, is inhibited by phosphorylation at Tyr(4473). Conversely, Shp2 binds most strongly when both of the NPXY motifs in LRP1 are phosphorylated.
Collapse
Affiliation(s)
- Gina N Betts
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093-0378, USA
| | | | | |
Collapse
|
47
|
Chen S, Bu G, Takei Y, Sakamoto K, Ikematsu S, Muramatsu T, Kadomatsu K. Midkine and LDL-receptor-related protein 1 contribute to the anchorage-independent cell growth of cancer cells. J Cell Sci 2007; 120:4009-15. [PMID: 17971413 DOI: 10.1242/jcs.013946] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The growth factor midkine (MK) is highly associated with cancer progression. Knockdown of MK expression strikingly suppresses tumor growth in nude mice. Thus, MK is a candidate target for cancer treatment. LDL-receptor-related protein 1 (LRP1) is a receptor for MK. We found that among the four ligand-binding domains of LRP1, the N-terminal half of the second domain (designated as MK-TRAP) had the strongest affinity to MK. MK-TRAP bound to MK, but not to HB-GAM/pleiotrophin, basic fibroblast growth factor or platelet-derived growth factor (PDGF)-BB. Exogenous MK-TRAP inhibited the binding between MK and LRP1. G401 cells that transiently or stably overexpress MK-TRAP showed decreased cell growth in monolayer culture and reduced colony formation in soft agar, which could be rescued by exogenous MK administration. MK-TRAP collected from conditioned medium also inhibited anchorage-independent growth of G401 cells and CMT-93 cells. Anti-MK antibody also inhibited the anchorage-independent growth. CMT-93 cells stably expressing MK-TRAP formed smaller tumors in a xenograft nude mouse model than control cells. Moreover, GST-RAP, a potent inhibitor of LRP1, inhibited the anchorage-independent growth of control G401 cells but not that of MK-TRAP stable transformants. Collectively, these data demonstrate a crucial role of MK-LRP1 signaling in anchorage-independent cell growth.
Collapse
Affiliation(s)
- Sen Chen
- Department of Biochemistry, Nagoya University School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | | | | | | | | | | | | |
Collapse
|
48
|
Minopoli G, Passaro F, Aloia L, Carlomagno F, Melillo RM, Santoro M, Forzati F, Zambrano N, Russo T. Receptor- and non-receptor tyrosine kinases induce processing of the amyloid precursor protein: role of the low-density lipoprotein receptor-related protein. NEURODEGENER DIS 2007; 4:94-100. [PMID: 17596703 DOI: 10.1159/000101833] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The Alzheimer's beta-amyloid peptides derive from the proteolytic processing of the beta-amyloid precursor protein, APP, by beta- and gamma-secretases. The regulation of this processing is not fully understood. Experimental evidence suggests that the activation of pathways involving protein tyrosine kinases, such as PDGFR and Src, could induce the cleavage of APP and in turn the generation of amyloid peptides. In this paper we addressed the effect of receptor and nonreceptor protein tyrosine kinases on the cleavage of APP and the mechanisms of their action. To this aim, we developed an in vitro system based on the APP-Gal4 fusion protein stably transfected in SHSY5Y neuroblastoma cell line. The cleavage of this molecule, induced by various stimuli, results in the activation of the transcription of the luciferase gene under the control of Gal4 cis-elements. By using this experimental system we demonstrated that, similarly to Src, three tyrosine kinases, TrkA, Ret and EGFR, induced the cleavage of APP-Gal4. We excluded that this effect was mediated by the activation of Ras-MAPK, PI3K-Akt and PLC-gamma pathways. Furthermore, the direct phosphorylation of the APP cytosolic domain does not affect Abeta peptide generation. On the contrary, experiments in cells lacking the LDL-receptor related protein LRP support the hypothesis that the interaction of APP with LRP is required for the induction of APP cleavage by tyrosine kinases.
Collapse
Affiliation(s)
- Giuseppina Minopoli
- CEINGE Biotecnologie Avanzate, Dipartimento di Biochimica e Biotecnologie Mediche, Università di Napoli Federico II, Napoli, Italia
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Salasznyk RM, Zappala M, Zheng M, Yu L, Wilkins-Port C, McKeown-Longo PJ. The uPA receptor and the somatomedin B region of vitronectin direct the localization of uPA to focal adhesions in microvessel endothelial cells. Matrix Biol 2007; 26:359-70. [PMID: 17344041 DOI: 10.1016/j.matbio.2007.01.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2006] [Revised: 01/26/2007] [Accepted: 01/29/2007] [Indexed: 01/26/2023]
Abstract
Vitronectin is a plasma protein which can deposit into the extracellular matrix where it supports integrin and uPA dependent cell migration. In earlier studies, we have shown that the plasma protein, vitronectin, stimulates focal adhesion remodeling by recruiting urokinase-type plasminogen activator (uPA) to focal adhesion sites [Wilcox-Adelman, S. A., Wilkins-Port, C. E., McKeown-Longo, P. J., 2000. Localization of urokinase-type plasminogen activator to focal adhesions requires ligation of vitronectin integrin receptors. Cell. Adhes. Commun.7, 477-490]. In the present study, we used a variety of vitronectin constructs to demonstrate that the localization of uPA to adhesion sites requires the binding of both vitronectin integrin receptors and the uPA receptor (uPAR) to vitronectin. A recombinant fragment of vitronectin containing the connecting sequence (VN(CS)) was able to support integrin-dependent adhesion, spreading and focal adhesion assembly by human microvessel endothelial cells. Cells adherent to this fragment were not able to localize uPA to focal adhesions. A second recombinant fragment containing both the amino-terminal SMB domain and the CS domain was able to restore the localization of uPA to adhesion sites. This fragment, which contains a uPAR binding site, also resulted in the localization of uPAR to adhesion sites. uPAR blocking antibodies as well as phospholipase C treatment of cells inhibited uPA localization to adhesion sites confirming a role for uPAR in this process. The SMB domain alone was unable to direct either uPAR or uPA to adhesion sites in the absence of the CS domain. Our results indicate that vitronectin-dependent localization of uPA to adhesion sites requires the sequential binding of vitronectin integrins and uPAR to vitronectin.
Collapse
Affiliation(s)
- Roman M Salasznyk
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New, Scotland Avenue, Albany, NY 12208, USA
| | | | | | | | | | | |
Collapse
|
50
|
Congote LF. Serpin A1 and CD91 as host instruments against HIV-1 infection: are extracellular antiviral peptides acting as intracellular messengers? Virus Res 2007; 125:119-34. [PMID: 17258834 DOI: 10.1016/j.virusres.2006.12.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2006] [Revised: 12/15/2006] [Accepted: 12/22/2006] [Indexed: 01/11/2023]
Abstract
Serpin A1 (alpha1-antitrypsin, alpha1-proteinase inhibitor) has been shown to be a non-cytolytic antiviral factor present in blood and effective against HIV infection. The best known physiological role of serpin A1 is to inhibit neutrophil elastase, a proteinase which is secreted by neutrophils at sites of infection and inflammation. Decreased HIV-infectivity is associated with decreased density of membrane-associated elastase. The enzyme may facilitate binding of the HIV membrane protein gp120 to host cells, and it specifically cleaves SDF-1, the physiological ligand of the HIV-1 co-receptor CXCR4. It has been suggested that one of the actions of serpin A1 as antiviral agent is to reduce HIV infectivity, and this property could be due to elastase inhibition. However, the most dramatic effect of serpin A1 is inhibition of HIV production. In vitro experiments indicate that the C-terminal peptide of serpin A1, produced during the formation of the complex of serpin with serine proteinases, may be responsible for the inhibition of HIV-1 expression in infected cells. This peptide, an integral part of the serpin-enzyme complex, is internalized by several scavenger receptors. Peptides corresponding to the C-terminal section of serpin A1 inhibit HIV-1 long-terminal-repeat-driven transcription and interact with nuclear proteins, such as alpha1-fetoprotein transcription factor. LDL-receptor-related protein 1 (LRP1/CD91), the best known receptor for serpin-enzyme complexes, is up-regulated in monocytes of HIV-1-infected true non-progressors. CD91 could be one of the major players in host resistance against HIV-1. It has the capacity of internalizing antiviral peptides such as serpin C-terminal fragments and alpha-defensins, and is at the same time the receptor for heat-shock proteins in antigen-presenting cells, in which chaperoned viral peptides could lead to the induction of cytotoxic T-cell responses.
Collapse
Affiliation(s)
- Luis Fernando Congote
- Endocrine Laboratory, McGill University Health Centre, 687 Avenue des pins, Ouest, Montreal, H3A 1A1, Canada.
| |
Collapse
|