1
|
Martin J, Falaise A, Faour S, Terryn C, Hachet C, Thiébault É, Huber L, Nizet P, Rioult D, Jaffiol R, Salesse S, Dedieu S, Langlois B. Differential Modulation of Endothelial Cell Functionality by LRP1 Expression in Fibroblasts and Cancer-Associated Fibroblasts via Paracrine signals and Matrix Remodeling. Matrix Biol 2025:S0945-053X(25)00048-4. [PMID: 40379110 DOI: 10.1016/j.matbio.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
LRP1 is a multifunctional endocytosis receptor involved in the regulation of cancer cell aggressiveness, fibroblast phenotype and angiogenesis. In breast cancer microenvironment, cancer-associated fibroblasts (CAFs) play a crucial role in matrix remodeling and tumor niche composition. LRP1 expression was described in fibroblasts and CAFs but remains poorly understood regarding its impact on endothelial cell behavior and angiocrine signaling. We analyzed the angio-modulatory effect of LRP1 expression in murine embryonic fibroblasts (MEFs) and breast cancer-educated CAF2 cells. We employed conditioned media and fibroblast-derived matrices to model fibroblastic cells angiogenic effects on human umbilical vein endothelial cells (HUVEC). Neither the extracellular matrix assembled by MEFs knock-out for LRP1 (PEA-13) nor their secretome modify the migration of HUVEC as compared to wild-type. Conversely, LRP1-deficient CAF2 secretome and matrices stimulate endothelial cell migration. Using spheroids, we demonstrate that PEA-13 secretome does not affect HUVEC angio-invasion. By contrast, CAF2 secretome invalidated for LRP1 stimulates endothelial sprouting as compared to controls. In addition, it specifically stabilized peripheral VE-cadherin-mediated endothelial cell junctions. A global proteomic analysis revealed that LRP1 expression in CAFs orchestrates a specific mobilization of secreted matricial components, surface receptors and membrane-associated proteins at the endothelial cell surface, thereby illustrating the deep influence exerted by LRP1 in angiogenic signals emitted by activated fibroblasts.
Collapse
Affiliation(s)
- Julie Martin
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France; Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Auréana Falaise
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France; Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Sara Faour
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France; Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France; Light, nanomaterials, nanotechnologies, ERL CNRS 7004, Université de Technologie de Troyes, Troyes, France
| | - Christine Terryn
- Plate-Forme Imagerie Cellulaire et Tissulaire (PICT), Université de Reims Champagne-Ardenne, UFR Médecine, Reims, France
| | - Cathy Hachet
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France; Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Émilie Thiébault
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France; Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Louise Huber
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France; Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Pierre Nizet
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France; Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Damien Rioult
- Plateau Technique Mobile de Cytométrie Environnementale MOBICYTE, Université de Reims Champagne-Ardenne/INERIS, Reims, France
| | - Rodolphe Jaffiol
- Light, nanomaterials, nanotechnologies, ERL CNRS 7004, Université de Technologie de Troyes, Troyes, France
| | - Stéphanie Salesse
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France; Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France.
| | - Stéphane Dedieu
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France; Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France.
| | - Benoit Langlois
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France; Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France.
| |
Collapse
|
2
|
Lee H, Aylward AJ, Pearse RV, Hsieh YC, Augur ZM, Benoit CR, Chou V, Knupp A, Pan C, Goberdhan S, Duong DM, Seyfried NT, Bennett DA, Klein HU, De Jager PL, Menon V, Young JE, Young-Pearse TL. Cell-type-specific regulation of APOE levels in human neurons by the Alzheimer's disease risk gene SORL1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.25.530017. [PMID: 36865313 PMCID: PMC9980168 DOI: 10.1101/2023.02.25.530017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
SORL1 is strongly implicated in the pathogenesis of Alzheimer's disease (AD) through human genetic studies that point to an association of reduced SORL1 levels with higher risk for AD. To interrogate the role(s) of SORL1 in human brain cells, SORL1 null iPSCs were generated, followed by differentiation to neuron, astrocyte, microglia, and endothelial cell fates. Loss of SORL1 led to alterations in both overlapping and distinct pathways across cell types, with the greatest effects in neurons and astrocytes. Intriguingly, SORL1 loss led to a dramatic neuron-specific reduction in APOE levels. Further, analyses of iPSCs derived from a human aging cohort revealed a neuron-specific linear correlation between SORL1 and APOE RNA and protein levels, a finding validated in human post-mortem brain. Pathway analysis implicated intracellular transport pathways and TGF- β/SMAD signaling in the function of SORL1 in neurons. In accord, enhancement of retromer-mediated trafficking and autophagy rescued elevated phospho-tau observed in SORL1 null neurons but did not rescue APOE levels, suggesting that these phenotypes are separable. Stimulation and inhibition of SMAD signaling modulated APOE RNA levels in a SORL1-dependent manner. These studies provide a mechanistic link between two of the strongest genetic risk factors for AD.
Collapse
|
3
|
Langlois B, Martin J, Schneider C, Hachet C, Terryn C, Rioult D, Martiny L, Théret L, Salesse S, Dedieu S. LRP-1-dependent control of calpain expression and activity: A new mechanism regulating thyroid carcinoma cell adhesion. Front Oncol 2022; 12:981927. [PMID: 36052226 PMCID: PMC9424861 DOI: 10.3389/fonc.2022.981927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
The low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional endocytic receptor mediating the clearance of various molecules from the extracellular matrix. LRP1 also regulates cell surface expression of matrix receptors by modulating both extracellular and intracellular signals, though current knowledge of the underlying mechanisms remains partial in the frame of cancer cells interaction with matricellular substrates. In this study we identified that LRP1 downregulates calpain activity and calpain 2 transcriptional expression in an invasive thyroid carcinoma cell model. LRP1-dependent alleviation of calpain activity limits cell-matrix attachment strength and contributes to FTC133 cells invasive abilities in a modified Boyden chamber assays. In addition, using enzymatic assays and co-immunoprecipitation experiments, we demonstrated that LRP1 exerts post-translational inhibition of calpain activity through PKA-dependent phosphorylation of calpain-2. This LRP-1 dual mode of control of calpain activity fine-tunes carcinoma cell spreading. We showed that LRP1-mediated calpain inhibition participates in talin-positive focal adhesions dissolution and limits β1-integrin expression at carcinoma cell surface. In conclusion, we identified an additional and innovative intracellular mechanism which demonstrates LRP-1 pro-motile action in thyroid cancer cells. LRP-1 ability to specifically control calpain-2 expression and activity highlights a novel facet of its de-adhesion receptor status.
Collapse
Affiliation(s)
- Benoit Langlois
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
- *Correspondence: Benoit Langlois,
| | - Julie Martin
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Christophe Schneider
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Cathy Hachet
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Christine Terryn
- Plate-Forme Imagerie Cellulaire et Tissulaire (PICT), Université de Reims Champagne-Ardenne, UFR Médecine, Reims, France
| | - Damien Rioult
- Plateau Technique Mobile de Cytométrie Environnementale MOBICYTE, Université de Reims Champagne-Ardenne/INERIS, Reims, France
| | - Laurent Martiny
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Louis Théret
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Stéphanie Salesse
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| | - Stéphane Dedieu
- UFR Sciences Exactes et Naturelles, Université de Reims Champagne-Ardenne, Reims, France
- Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, UMR 7369 CNRS, Reims, France
| |
Collapse
|
4
|
Cai H, Cai T, Zheng H, Liu L, Zhou L, Pang X, Zhan Q, Wang Y, Yang C, Guo Z, Pan H, Wang Q. The Neuroprotective Effects of Danggui-Shaoyao San on Vascular Cognitive Impairment: Involvement of the Role of the Low-Density Lipoprotein Receptor-Related Protein. Rejuvenation Res 2020; 23:420-433. [PMID: 32242481 DOI: 10.1089/rej.2019.2182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Haobin Cai
- Department of Neurology & Psychology, Shenzhen Traditional Chinese Medicine Hospital, Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Tiantian Cai
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haotao Zheng
- Department of Neurology & Psychology, Shenzhen Traditional Chinese Medicine Hospital, Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lijin Liu
- Department of Neurology & Psychology, Shenzhen Traditional Chinese Medicine Hospital, Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Liuchang Zhou
- Department of Neurology & Psychology, Shenzhen Traditional Chinese Medicine Hospital, Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xile Pang
- Department of Neurology & Psychology, Shenzhen Traditional Chinese Medicine Hospital, Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Qinkai Zhan
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yijie Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cong Yang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhouke Guo
- Department of Neurology & Psychology, Shenzhen Traditional Chinese Medicine Hospital, Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Huafeng Pan
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
5
|
Zhang M, Ding X, Zhang Q, Liu J, Zhang Y, Zhang Y, Tian Z, Li W, Zhu W, Kang H, Wang Z, Wu X, Wang C, Yang X, Wang K. Exome sequencing of 112 trios identifies recessive genetic variants in brain arteriovenous malformations. J Neurointerv Surg 2020; 13:568-573. [PMID: 32848021 DOI: 10.1136/neurintsurg-2020-016469] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 02/02/2023]
Abstract
BACKGROUND Brain arteriovenous malformation (BAVM) is a main cause of cerebral hemorrhage and hemorrhagic stroke in adolescents. Morphologically, a BAVM is an abnormal connection between cerebrovascular arteries and veins. The genetic etiology of BAVMs has not been fully elucidated. In this study, we aim to investigate potential recessive genetic variants in BAVMs by interrogation of rare compound heterozygous variants. METHODS We performed whole exome sequencing (WES) on 112 BAVM trios and analyzed the data for rare and deleterious compound heterozygous mutations associated with the disease. RESULTS We identified 16 genes with compound heterozygous variants that were recurrent in more than one trio. Two genes (LRP2, MUC5B) were recurrently mutated in three trios. LRP2 has been previously associated with BAVM pathogenesis. Fourteen genes (MYLK, HSPG2, PEAK1, PIEZO1, PRUNE2, DNAH14, DNAH5, FCGBP, HERC2, HMCN1, MYH1, NHSL1, PLEC, RP1L1) were recurrently mutated in two trios, and five of these genes (MYLK, HSPG2, PEAK1, PIEZO1, PRUNE2) have been reported to play a role in angiogenesis or vascular diseases. Additionally, abnormal expression of the MYLK protein is related to spinal arteriovenous malformations. CONCLUSION Our study indicates that rare recessive compound heterozygous variants may underlie cases of BAVM. These findings improve our understanding of BAVM pathology and indicate genes for functional validation.
Collapse
Affiliation(s)
- Mingqi Zhang
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Xinghuan Ding
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing 100070, China
| | - Qianqian Zhang
- Department of Cerebrovascular Disease, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Henan Provincial Neurointerventional Engineering Research Center and Henan International Joint Laboratory of Cerebrovascular Disease, Zhengzhou 450000, Henan, China
| | - Jian Liu
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Yisen Zhang
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Ying Zhang
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Zhongbin Tian
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Wenqiang Li
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Wei Zhu
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Huibin Kang
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Zhongxiao Wang
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Xinzhi Wu
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Chao Wang
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Xinjian Yang
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Kun Wang
- Department of Interventional Neuroradiology, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| |
Collapse
|
6
|
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: 44] [Impact Index Per Article: 7.3] [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
|
7
|
Vinik Y, Shatz-Azoulay H, Vivanti A, Hever N, Levy Y, Karmona R, Brumfeld V, Baraghithy S, Attar-Lamdar M, Boura-Halfon S, Bab I, Zick Y. The mammalian lectin galectin-8 induces RANKL expression, osteoclastogenesis, and bone mass reduction in mice. eLife 2015; 4:e05914. [PMID: 25955862 PMCID: PMC4424493 DOI: 10.7554/elife.05914] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 04/16/2015] [Indexed: 01/20/2023] Open
Abstract
Skeletal integrity is maintained by the co-ordinated activity of osteoblasts, the bone-forming cells, and osteoclasts, the bone-resorbing cells. In this study, we show that mice overexpressing galectin-8, a secreted mammalian lectin of the galectins family, exhibit accelerated osteoclasts activity and bone turnover, which culminates in reduced bone mass, similar to cases of postmenopausal osteoporosis and cancerous osteolysis. This phenotype can be attributed to a direct action of galectin-8 on primary cultures of osteoblasts that secrete the osteoclastogenic factor RANKL upon binding of galectin-8. This results in enhanced differentiation into osteoclasts of the bone marrow cells co-cultured with galectin-8-treated osteoblasts. Secretion of RANKL by galectin-8-treated osteoblasts can be attributed to binding of galectin-8 to receptor complexes that positively (uPAR and MRC2) and negatively (LRP1) regulate galectin-8 function. Our findings identify galectins as new players in osteoclastogenesis and bone remodeling, and highlight a potential regulation of bone mass by animal lectins. DOI:http://dx.doi.org/10.7554/eLife.05914.001 The forces applied to the body during daily activities cause bones to be constantly remodeled, which is essential for keeping them healthy. In most adult organisms, new bone is created at the same rate at which old bone is destroyed. This means that overall bone mass remains the same. But, in diseases such as osteoporosis or bone cancer, bone is destroyed more rapidly than at which new bone is made. This leads to brittle bones that are more likely to fracture. Understanding how to increase the rate of bone renewal might therefore help scientists develop new treatments for bone diseases. Bone is created by cells called osteoblasts and destroyed by other cells called osteoclasts. Both of these types of cells develop from stem cells in the bone marrow. The activity of these cells is controlled by a number of factors, including the matrix of proteins that holds bone together. A group of proteins called galectins are known to act as a bridge between some of the matrix proteins and molecules on the surface of the cells. Vinik et al. took osteoblasts from a mouse skull, grew them in the laboratory, and then exposed them to a galectin protein called galectin-8. This made the osteoblasts release a protein called RANKL, which is known to boost osteoclast activity. When osteoblasts that had been exposed to galectin-8 were grown alongside bone marrow stem cells, more of the stem cells developed into the bone-destroying osteoclasts. Mice that were genetically engineered to produce more galectin-8 than normal mice develop brittle bones, despite also creating new bone at a higher rate than do normal mice. This is because osteoclast activity increases at a greater rate, resulting in an overall loss of bone in these animals. This is similar to what occurs in some individuals with osteoporosis. These experiments therefore suggest that galectin-8 plays an important role in bone remodeling and that it may be a potential target for drugs that treat diseases that weaken bones. DOI:http://dx.doi.org/10.7554/eLife.05914.002
Collapse
Affiliation(s)
- Yaron Vinik
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hadas Shatz-Azoulay
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Alessia Vivanti
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Navit Hever
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yifat Levy
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Rotem Karmona
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Vlad Brumfeld
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Saja Baraghithy
- Bone Laboratory, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Sigalit Boura-Halfon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Itai Bab
- Bone Laboratory, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yehiel Zick
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
8
|
Strickland DK, Au DT, Cunfer P, Muratoglu SC. Low-density lipoprotein receptor-related protein-1: role in the regulation of vascular integrity. Arterioscler Thromb Vasc Biol 2014; 34:487-98. [PMID: 24504736 DOI: 10.1161/atvbaha.113.301924] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Low-density lipoprotein receptor-related protein-1 (LRP1) is a large endocytic and signaling receptor that is widely expressed. In the liver, LRP1 plays an important role in regulating the plasma levels of blood coagulation factor VIII (fVIII) by mediating its uptake and subsequent degradation. fVIII is a key plasma protein that is deficient in hemophilia A and circulates in complex with von Willebrand factor. Because von Willebrand factor blocks binding of fVIII to LRP1, questions remain on the molecular mechanisms by which LRP1 removes fVIII from the circulation. LRP1 also regulates cell surface levels of tissue factor, a component of the extrinsic blood coagulation pathway. This occurs when tissue factor pathway inhibitor bridges the fVII/tissue factor complex to LRP1, resulting in rapid LRP1-mediated internalization and downregulation of coagulant activity. In the vasculature LRP1 also plays protective role from the development of aneurysms. Mice in which the lrp1 gene is selectively deleted in vascular smooth muscle cells develop a phenotype similar to the progression of aneurysm formation in human patient, revealing that these mice are ideal for investigating molecular mechanisms associated with aneurysm formation. Studies suggest that LRP1 protects against elastin fiber fragmentation by reducing excess protease activity in the vessel wall. These proteases include high-temperature requirement factor A1, matrix metalloproteinase 2, matrix metalloproteinase-9, and membrane associated type 1-matrix metalloproteinase. In addition, LRP1 regulates matrix deposition, in part, by modulating levels of connective tissue growth factor. Defining pathways modulated by LRP1 that lead to aneurysm formation and defining its role in thrombosis may allow for more effective intervention in patients.
Collapse
Affiliation(s)
- Dudley K Strickland
- From the Center for Vascular and Inflammatory Disease (D.K.S., D.T.A., P.C., S.C.M.), Departments of Surgery (D.K.S.), and Physiology (S.C.M.), University of Maryland School of Medicine, Baltimore
| | | | | | | |
Collapse
|
9
|
Sauer H, Ravindran F, Beldoch M, Sharifpanah F, Jedelská J, Strehlow B, Wartenberg M. α2-Macroglobulin enhances vasculogenesis/angiogenesis of mouse embryonic stem cells by stimulation of nitric oxide generation and induction of fibroblast growth factor-2 expression. Stem Cells Dev 2013; 22:1443-54. [PMID: 23379699 DOI: 10.1089/scd.2012.0640] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
α2-macroglobulin (α2M) is an acute-phase protein released upon challenges like cardiac hypertrophy and infarction. α2M signals via the low density lipoprotein receptor-related protein (LRP-1) and may induce stem cell activation. In the present study, the effects of α2M on vasculogenesis/angiogenesis and underlying signaling cascades were investigated in mouse embryonic stem (ES) cells. LRP-1 was expressed in ES cells and upregulated during differentiation. α2M dose dependently increased CD31-positive vascular structures in ES cell-derived embryoid bodies, the early cardiovascular markers isl-1, Nkx-2.5, and flk-1 as well as numbers of VE-cadherin and flk-1-positive cells, but downregulated α-smooth muscle actin. Enhancement of vasculogenesis/angiogenesis by α2M was abolished by the LRP-1 antagonist receptor-associated protein (RAP) and LRP-1 blocking antibody. Notably, α2M stimulated vascular growth in the chicken chorioallantois membrane assay, but not in a human umbilical vein endothelial cell spheroid model. α2M increased fibroblast growth factor-2 (FGF-2) protein expression, which was abolished by RAP, induced nitric oxide (NO) generation as determined by 4,5-diaminofluorescein diacetate microfluorometry, and activated nitric oxide synthase-3 (NOS-3) as well as extracellular-regulated kinase 1,2 (ERK1/2) and phosphatidyl inositol 3-kinase (PI3K). NO generation, the increase in FGF-2 expression, and the stimulation of vasculogenesis/angiogenesis by α2M were blunted by the NO synthase inhibitor L-NAME, the ERK1/2 inhibitor PD98059, and the PI3K inhibitor LY294002. Furthermore, vasculogenesis/angiogenesis by α2M was inhibited in the presence of the FGF receptor 1 antagonist SU5402. In conclusion, α2M stimulates endothelial and early cardiac, but not smooth muscle differentiation of ES cells through generation of NO, activation of ERK1/2 as well as PI3K, and induction of FGF-2 expression.
Collapse
Affiliation(s)
- Heinrich Sauer
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany.
| | | | | | | | | | | | | |
Collapse
|
10
|
Costales P, Castellano J, Revuelta-López E, Cal R, Aledo R, Llampayas O, Nasarre L, Juarez C, Badimon L, Llorente-Cortés V. Lipopolysaccharide downregulates CD91/low-density lipoprotein receptor-related protein 1 expression through SREBP-1 overexpression in human macrophages. Atherosclerosis 2012; 227:79-88. [PMID: 23312784 DOI: 10.1016/j.atherosclerosis.2012.12.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 12/11/2012] [Accepted: 12/14/2012] [Indexed: 12/18/2022]
Abstract
Sterol regulatory element-binding proteins (SREBPs) negatively modulate the expression of the CD91/low-density lipoprotein receptor-related protein (LRP1), a carrier and signaling receptor that mediates the endocytosis of more than 40 structurally and functionally distinct ligands. The aim of this work was to analyze whether lipopolysaccharide (LPS) can regulate LRP1 expression through SREBPs in human monocyte-derived macrophages (HMDM). LPS led to LRP1 mRNA and protein inhibition in a dose- and time-dependent manner. Concomitantly, a strong upregulation of SREBP-1 mRNA and SREBP-1 nuclear protein levels was observed in LPS-treated HMDM. The specific silencing of SREBP-1 efficiently prevented LRP1 reduction caused by LPS. SREBP-1 mRNA and nuclear protein levels remained high in HMDM treated with LPS unexposed or exposed to LDL. Native (nLDL) or aggregated LDL (agLDL) per se downregulated SREBP-2 expression levels and increased LRP1 expression. However, lipoproteins did not significantly alter the effect of LPS on SREBP-1 and LRP1 expression. Collectively, these data support that lipoproteins and LPS exert their modulatory effect on LRP1 expression through different SREBP isoforms, SREBP-2 and SREBP-1, respectively. These results highlight a crucial role of SREBP-1 as a mediator of the downregulatory effects of LPS on LRP1 expression in human macrophages, independently of the absence or presence of modified lipoproteins.
Collapse
Affiliation(s)
- P Costales
- Cardiovascular Research Center, CSIC-ICCC, IIB-Sant Pau, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Song P, Bao H, Yu Y, Xue Y, Yun D, Zhang Y, He Y, Liu Y, Liu Q, Lu H, Fan H, Luo J, Yang P, Chen X. Comprehensive profiling of metastasis-related proteins in paired hepatocellular carcinoma cells with different metastasis potentials. Proteomics Clin Appl 2012; 3:841-52. [PMID: 21136991 DOI: 10.1002/prca.200780131] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Precise and comprehensive identifications of the proteins associated with metastasis are critical for early diagnosis and therapeutic intervention of hepatocellular carcinoma (HCC). Therefore, we investigated the proteomic differences between a pair of HCC cell lines, originating from the same progenitor, with different metastasis potential using amino acid-coded mass tagging-based LC-MS/MS quantitative proteomic approach. Totally the relative abundance of 336 proteins in these cell lines were quantified, in which 121 proteins were upregulated by >30%, and 64 proteins were downregulated by >23% in the cells with high metastasis potential. Further validation studies by Western blotting in a series of HCC cell types with progressively increasing trend of metastasis showed that peroxiredoxin 4, HSP90β and HSP27 were positively correlated with increasing metastasis while prohibitin was negatively correlated with metastasis potential. These validation results were also consistent with that obtained from comparative analysis of clinic tissues samples. Function annotations of differentially expressed HCC proteome suggested that the emergence and development of high metastasis involved the dysregulation of cell migration, cell cycle and membrane traffics. Together our results revealed a much more comprehensive profile than that from 2-DE-based method and provided more global insights into the mechanisms of HCC metastasis and potential markers for clinical diagnosis.
Collapse
Affiliation(s)
- Peiming Song
- College of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, P. R. China; Institutes of Biomedical Science, Fudan University, Shanghai, P. R. China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Llorente-Cortes V, Casani L, Cal R, Llenas A, Juan-Babot O, Camino-López S, Sendra J, Badimon L. Cholesterol-lowering strategies reduce vascular LRP1 overexpression induced by hypercholesterolaemia. Eur J Clin Invest 2011; 41:1087-97. [PMID: 21434892 DOI: 10.1111/j.1365-2362.2011.02513.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Low density lipoprotein receptor-related protein (LRP1) plays a key role on vascular functionality and is upregulated by hypercholesterolemia and hypertension. To investigate the effect of cholesterol-lowering interventions on vascular LRP1 over expression and whether simvastatin influences LRP1 expression. MATERIAL AND METHODS Male New Zealand rabbits were recruited into various groups, one group was fed a normal chow diet for 28 days (control group, n = 6), other group (n = 24) was fed a hypercholesterolemic diet (HC), six rabbits were euthanized at day 28 to test the capacity of HC diet to induce early atherosclerosis and the rest at day 60 (n = 18) after receiving either HC diet (HC group, n = 6), HC diet with simvastatin (2·5 mg/kg.day) (HC+simv group, n = 6), or a normal chow diet (NC group, n = 6) for the last 32 days. RESULTS High-cholesterol diet raised vascular LRP1 concomitantly with increased lipid, VSMC and macrophage content in the arterial intima. Simvastatin and return to normocholesterolemic diet significantly reduced systemic cholesterol levels and vascular lipid content. Interestingly, these interventions also downregulate LRP1 overexpression in the vascular wall although to a different extent (HC+simv: 75 ± 3·6%vs NC: 50 ± 3·5% versus, P = 0·002). Immunohistochemistry studies showed that LRP1 diminushion was associated to a reduction in the number of intimal VSMC in HC+simv.group. Simvastatin per se did not exert any significant effect on LRP1 expression in rabbit aortic smooth muscle cells (rSMC). CONCLUSIONS Our results demonstrate that cholesterol-lowering interventions exerted down regulatory effects on vascular LRP1 over expression induced by hypercholesterolemia and that simvastatin did not influence LRP1 expression beyond its cholesterol-lowering effects.
Collapse
Affiliation(s)
- Vicenta Llorente-Cortes
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau-UAB, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Burns JS, Kristiansen M, Kristensen LP, Larsen KH, Nielsen MO, Christiansen H, Nehlin J, Andersen JS, Kassem M. Decellularized matrix from tumorigenic human mesenchymal stem cells promotes neovascularization with galectin-1 dependent endothelial interaction. PLoS One 2011; 6:e21888. [PMID: 21779348 PMCID: PMC3133605 DOI: 10.1371/journal.pone.0021888] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 06/13/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Acquisition of a blood supply is fundamental for extensive tumor growth. We recently described vascular heterogeneity in tumours derived from cell clones of a human mesenchymal stem cell (hMSC) strain (hMSC-TERT20) immortalized by retroviral vector mediated human telomerase (hTERT) gene expression. Histological analysis showed that cells of the most vascularized tumorigenic clone, -BD11 had a pericyte-like alpha smooth muscle actin (ASMA+) and CD146+ positive phenotype. Upon serum withdrawal in culture, -BD11 cells formed cord-like structures mimicking capillary morphogenesis. In contrast, cells of the poorly tumorigenic clone, -BC8 did not stain for ASMA, tumours were less vascularized and serum withdrawal in culture led to cell death. By exploring the heterogeneity in hMSC-TERT20 clones we aimed to understand molecular mechanisms by which mesenchymal stem cells may promote neovascularization. METHODOLOGY/PRINCIPAL FINDINGS Quantitative qRT-PCR analysis revealed similar mRNA levels for genes encoding the angiogenic cytokines VEGF and Angiopoietin-1 in both clones. However, clone-BD11 produced a denser extracellular matrix that supported stable ex vivo capillary morphogenesis of human endothelial cells and promoted in vivo neovascularization. Proteomic characterization of the -BD11 decellularized matrix identified 50 extracellular angiogenic proteins, including galectin-1. siRNA knock down of galectin-1 expression abrogated the ex vivo interaction between decellularized -BD11 matrix and endothelial cells. More stable shRNA knock down of galectin-1 expression did not prevent -BD11 tumorigenesis, but greatly reduced endothelial migration into -BD11 cell xenografts. CONCLUSIONS Decellularized hMSC matrix had significant angiogenic potential with at least 50 angiogenic cell surface and extracellular proteins, implicated in attracting endothelial cells, their adhesion and activation to form tubular structures. hMSC -BD11 surface galectin-1 expression was required to bring about matrix-endothelial interactions and for xenografted hMSC -BD11 cells to optimally recruit host vasculature.
Collapse
Affiliation(s)
- Jorge S Burns
- Molecular Endocrinology Laboratory KMEB, Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
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 PMCID: PMC2997161 DOI: 10.1021/pr1008288] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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, CA, 92093
| | - Gabriel Simon
- Department of Chemical Biology and the Skaggs Institute for Chemical Biology, La Jolla, CA, 92093
| | - Sherry Niessen
- Department of Chemical Biology and the Skaggs Institute for Chemical Biology, La Jolla, CA, 92093
- Center for Physiological Proteomics, Scripps Research institute, La Jolla, CA, 92093
| | - Melissa Dix
- Department of Chemical Biology and the Skaggs Institute for Chemical Biology, La Jolla, CA, 92093
| | - Shinako Takimoto
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA, 92093
| | - Benjamin F. Cravatt
- Department of Chemical Biology and the Skaggs Institute for Chemical Biology, La Jolla, CA, 92093
| | - Steven L. Gonias
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA, 92093
| |
Collapse
|
15
|
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
|
16
|
Gaultier A, Hollister M, Reynolds I, Hsieh EH, Gonias SL. LRP1 regulates remodeling of the extracellular matrix by fibroblasts. Matrix Biol 2010; 29:22-30. [PMID: 19699300 PMCID: PMC2818094 DOI: 10.1016/j.matbio.2009.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 08/04/2009] [Accepted: 08/11/2009] [Indexed: 10/25/2022]
Abstract
Low density lipoprotein receptor-related protein (LRP1) is an endocytic receptor for diverse proteases, protease inhibitors, and other plasma membrane proteins, including the urokinase receptor (uPAR). LRP1 also functions in cell-signaling and regulates gene expression. The goal of this study was to determine whether LRP1 regulates remodeling of provisional extracellular matrix (ECM) by fibroblasts. To address this problem, we utilized an in vitro model in which type I collagen was reconstituted and overlaid with fibronectin. Either the collagen or fibronectin was fluorescently-labeled. ECM remodeling by fibroblasts deficient in LRP1, uPAR, or MT1-MMP was studied. MT1-MMP was required for efficient remodeling of the deep collagen layer but not involved in fibronectin remodeling. Instead, fibronectin was remodeled by a system that required urokinase-type plasminogen activator (uPA), uPAR, and exogenously-added plasminogen. LRP1 markedly inhibited fibronectin remodeling by regulating cell-surface uPAR and plasminogen activation. LRP1 also regulated remodeling of the deep collagen layer but not by controlling MT1-MMP. Instead, LRP1 deficiency or inhibition de-repressed a secondary pathway for collagen remodeling, which was active in MT1-MMP-deficient cells but not in uPAR-deficient cells. These results demonstrate that LRP1 regulates ECM remodeling principally by repressing pathways that require plasminogen activation by uPA in association with uPAR.
Collapse
Affiliation(s)
- Alban Gaultier
- Department of Pathology, University of California San Diego, La Jolla, California 92093, USA
| | | | | | | | | |
Collapse
|
17
|
Camino-López S, Badimon L, González A, Canals D, Peña E, Llorente-Cortés V. Aggregated low density lipoprotein induces tissue factor by inhibiting sphingomyelinase activity in human vascular smooth muscle cells. J Thromb Haemost 2009; 7:2137-46. [PMID: 19817993 DOI: 10.1111/j.1538-7836.2009.03638.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Our previous results demonstrated that aggregated low density lipoprotein (agLDL) induces tissue factor (TF) expression and activation through Rho A translocation in human vascular smooth muscle cells (VSMC). We also previously demonstrated that membrane sphingomyelin (SM) content is higher in agLDL-exposed VSMC than in control cells. The main enzymes regulating cellular SM content are the family of sphingomyelinases (Smases) that hydrolize SM to phosphorylcholine and ceramide (CER). OBJECTIVES We wished to investigate whether agLDL has the ability to modulate acidic- (A-) and neutral (N-) Smase activity and whether or not this effect is related to the upregulatory effect of agLDL on Rho A translocation and TF activation in human VSMC. METHODS AND RESULTS By measuring generated [(14)C]-phosphorylcholine, we found that agLDL significantly decreased A-Smase and specially N-Smase activity. Pharmacological Smase inhibitors increased Rho A and TF. Specific loss-of-function of A-Smase or N-Smase 1 (N1-Smase) by siRNA treatment (500 nmol L(-1), 12 hours) dramatically increased membrane Rho A protein levels (5- and 3-fold, respectively). Concomitantly, TF protein expression and TF procoagulant activity were also increased. Inhibition of A-Smase or N-Smase activity by agLDL, siRNA-anti A- or N1-Smase or pharmacological treatment significantly increased the SM content of vascular cells. The inhibition of SM synthesis by fumonisin B(1) (FB(1)) prevented the upregulatory effect of agLDL on TF. CONCLUSIONS These results demonstrate that inhibition of both A- and N1-Smase might explain the upregulatory effect of agLDL on TF activation, and suggest that this effect is related, at least in part, to membrane SM enrichment.
Collapse
Affiliation(s)
- S Camino-López
- Cardiovascular Research Center of Barcelona, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, Barcelona
| | | | | | | | | | | |
Collapse
|
18
|
Mace KA, Restivo TE, Rinn JL, Paquet AC, Chang HY, Young DM, Boudreau NJ. HOXA3 modulates injury-induced mobilization and recruitment of bone marrow-derived cells. Stem Cells 2009; 27:1654-65. [PMID: 19544454 PMCID: PMC2733377 DOI: 10.1002/stem.90] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulated recruitment and differentiation of multipotent bone marrow-derived cells (BMDCs) to sites of injury are critical for efficient wound healing. Previously we demonstrated that sustained expression of HOXA3 both accelerated wound healing and promoted angiogenesis in diabetic mice. In this study, we have used green fluorescent protein-positive bone marrow chimeras to investigate the effect of HOXA3 expression on recruitment of BMDCs to wounds. We hypothesized that the enhanced neovascularization induced by HOXA3 is due to enhanced mobilization, recruitment, and/or differentiation of BMDCs. Here we show that diabetic mice treated with HOXA3 displayed a significant increase in both mobilization and recruitment of endothelial progenitor cells compared with control mice. Importantly, we also found that HOXA3-treated mice had significantly fewer inflammatory cells recruited to the wound compared with control mice. Microarray analyses of HOXA3-treated wounds revealed that indeed HOXA3 locally increased expression of genes that selectively promote stem/progenitor cell mobilization and recruitment while also suppressing expression of numerous members of the proinflammatory nuclear factor κB pathway, including myeloid differentiation primary response gene 88 and toll-interacting protein. Thus HOXA3 accelerates wound repair by mobilizing endothelial progenitor cells and attenuating the excessive inflammatory response of chronic wounds.
Collapse
Affiliation(s)
- Kimberly A Mace
- Department of Surgery, University of California San Francisco, San Francisco, California 94143-1302, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
Dakhova O, Ozen M, Creighton CJ, Li R, Ayala G, Rowley D, Ittmann M. Global gene expression analysis of reactive stroma in prostate cancer. Clin Cancer Res 2009; 15:3979-89. [PMID: 19509179 PMCID: PMC2734921 DOI: 10.1158/1078-0432.ccr-08-1899] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE Marked reactive stroma formation, designated as grade 3 reactive stroma, is associated with poor outcome in clinically localized prostate cancer. To understand the biological processes and signaling mechanisms underlying the formation of such reactive stroma, we carried out microarray gene expression analysis of laser-captured reactive stroma and matched normal stroma. EXPERIMENTAL DESIGN Seventeen cases of reactive stroma grade 3 cancer were used to laser-capture tumor and normal stroma. Expression analysis was carried out using Agilent 44K arrays. Up-regulation of selected genes was confirmed by quantitative reverse transcription-PCR. Expression data was analyzed to identify significantly up- and down-regulated genes, and gene ontology analysis was used to define pathways altered in reactive stroma. RESULTS A total of 544 unique genes were significantly higher in the reactive stroma and 606 unique genes were lower. Gene ontology analysis revealed significant alterations in a number of novel processes in prostate cancer reactive stroma, including neurogenesis, axonogenesis, and the DNA damage/repair pathways, as well as evidence of increases in stem cells in prostate cancer reactive stroma. CONCLUSIONS Formation of reactive stroma in prostate cancer is a dynamic process characterized by significant alterations in growth factor and signal transduction pathways and formation of new structures, including nerves and axons.
Collapse
Affiliation(s)
- Olga Dakhova
- Dept. of Pathology, Baylor College of Medicine, Houston, Texas 77030
- Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas 77030
| | - Mustafa Ozen
- Dept. of Pathology, Baylor College of Medicine, Houston, Texas 77030
- Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas 77030
| | | | - Rile Li
- Dept. of Pathology, Baylor College of Medicine, Houston, Texas 77030
| | - Gustavo Ayala
- Dept. of Pathology, Baylor College of Medicine, Houston, Texas 77030
| | - David Rowley
- Dept. of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Michael Ittmann
- Dept. of Pathology, Baylor College of Medicine, Houston, Texas 77030
- Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas 77030
| |
Collapse
|
20
|
Dedieu S, Langlois B. LRP-1: a new modulator of cytoskeleton dynamics and adhesive complex turnover in cancer cells. Cell Adh Migr 2009; 2:77-80. [PMID: 19271352 DOI: 10.4161/cam.2.2.6374] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The low-density lipoprotein receptor-related protein-1 (LRP-1)is a large scavenger receptor mediating the internalization and catabolism of various biological components from the extracellular matrix. In the past decade, LRP-1 appeared as an attractive receptor for targeting the invasive behavior of cancer cells since this protein is able to reduce the accumulation of extracellular proteinases by endocytosis. However, recent data suggest that LRP-1 could support carcinoma cell invasion depending on the cellular environment. Indeed, in addition to its well-determined role in ligand binding and endocytosis, LRP-1 emerges as a central molecular regulator of cytoskeleton organization and adhesive complex turnover in malignant cells. This commentary reviews the functions played by LRP-1 in cancer-related events and discusses the potential mechanisms whereby LRP-1 is able to control the cellular phenotype of cancer cells.
Collapse
Affiliation(s)
- Stéphane Dedieu
- Université de Reims Champagne-Ardenne (URCA), CNRS UMR MEDyC 6237, Laboratoire SiRMa, Reims, France.
| | | |
Collapse
|
21
|
Song PM, Zhang Y, He YF, Bao HM, Luo JH, Liu YK, Yang PY, Chen X. Bioinformatics analysis of metastasis-related proteins in hepatocellular carcinoma. World J Gastroenterol 2008; 14:5816-22. [PMID: 18855979 PMCID: PMC2751890 DOI: 10.3748/wjg.14.5816] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To analyze the metastasis-related proteins in hepatocellular carcinoma (HCC) and discover the biomarker candidates for diagnosis and therapeutic intervention of HCC metastasis with bioinformatics tools.
METHODS: Metastasis-related proteins were determined by stable isotope labeling and MS analysis and analyzed with bioinformatics resources, including Phobius, Kyoto encyclopedia of genes and genomes (KEGG), online mendelian inheritance in man (OMIM) and human protein reference database (HPRD).
RESULTS: All the metastasis-related proteins were linked to 83 pathways in KEGG, including MAPK and p53 signal pathways. Protein-protein interaction network showed that all the metastasis-related proteins were categorized into 19 function groups, including cell cycle, apoptosis and signal transduction. OMIM analysis linked these proteins to 186 OMIM entries.
CONCLUSION: Metastasis-related proteins provide HCC cells with biological advantages in cell proliferation, migration and angiogenesis, and facilitate metastasis of HCC cells. The bird’s eye view can reveal a global characteristic of metastasis-related proteins and many differentially expressed proteins can be identified as candidates for diagnosis and treatment of HCC.
Collapse
|
22
|
Gaultier A, Arandjelovic S, Niessen S, Overton CD, Linton MF, Fazio S, Campana WM, Cravatt BF, Gonias SL. Regulation of tumor necrosis factor receptor-1 and the IKK-NF-kappaB pathway by LDL receptor-related protein explains the antiinflammatory activity of this receptor. Blood 2008; 111:5316-25. [PMID: 18369152 PMCID: PMC2396725 DOI: 10.1182/blood-2007-12-127613] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Accepted: 03/18/2008] [Indexed: 12/18/2022] Open
Abstract
Low-density lipoprotein receptor-related protein (LRP-1) functions in endocytosis and in cell signaling directly (by binding signaling adaptor proteins) or indirectly (by regulating levels of other cell-surface receptors). Because recent studies in rodents suggest that LRP-1 inhibits inflammation, we conducted activity-based protein profiling experiments to discover novel proteases, involved in inflammation, that are regulated by LRP-1. We found that activated complement proteases accumulate at increased levels when LRP-1 is absent. Although LRP-1 functions as an endocytic receptor for C1r and C1s, complement protease mRNA expression was increased in LRP-1-deficient cells, as was expression of inducible nitric oxide synthase (iNOS) and interleukin-6. Regulation of expression of inflammatory mediators was explained by the ability of LRP-1 to suppress basal cell signaling through the I kappaB kinase-nuclear factor-kappaB (NF-kappaB) pathway. LRP-1-deficient macrophages, isolated from mice, demonstrated increased expression of iNOS, C1r, and monocyte chemoattractant protein-1 (MCP-1); MCP-1 expression was inhibited by NF-kappaB antagonism. The mechanism by which LRP-1 inhibits NF-kappaB activity involves down-regulating cell-surface tumor necrosis factor receptor-1 (TNFR1) and thus, inhibition of autocrine TNFR1-initiated cell signaling. TNF-alpha-neutralizing antibody inhibited NF-kappaB activity selectively in LRP-1-deficient cells. We propose that LRP-1 suppresses expression of inflammatory mediators indirectly, by regulating TNFR1-dependent cell signaling through the I kappaB kinase-NF-kappaB pathway.
Collapse
Affiliation(s)
- Alban Gaultier
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA 92093-0612, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Gaultier A, Arandjelovic S, Li X, Janes J, Dragojlovic N, Zhou GP, Dolkas J, Myers RR, Gonias SL, Campana WM. A shed form of LDL receptor-related protein-1 regulates peripheral nerve injury and neuropathic pain in rodents. J Clin Invest 2008; 118:161-72. [PMID: 18060043 DOI: 10.1172/jci32371] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Accepted: 10/03/2007] [Indexed: 12/11/2022] Open
Abstract
Injury to the peripheral nervous system (PNS) initiates a response controlled by multiple extracellular mediators, many of which contribute to the development of neuropathic pain. Schwann cells in an injured nerve demonstrate increased expression of LDL receptor-related protein-1 (LRP1), an endocytic receptor for diverse ligands and a cell survival factor. Here we report that a fragment of LRP1, in which a soluble or shed form of LRP1 with an intact alpha-chain (sLRP-alpha), was shed by Schwann cells in vitro and in the PNS after injury. Injection of purified sLRP-alpha into mouse sciatic nerves prior to chronic constriction injury (CCI) inhibited p38 MAPK activation (P-p38) and decreased expression of TNF-alpha and IL-1beta locally. sLRP-alpha also inhibited CCI-induced spontaneous neuropathic pain and decreased inflammatory cytokine expression in the spinal dorsal horn, where neuropathic pain processing occurs. In cultures of Schwann cells, astrocytes, and microglia, sLRP-alpha inhibited TNF-alpha-induced activation of p38 MAPK and ERK/MAPK. The activity of sLRP-alpha did not involve TNF-alpha binding, but rather glial cell preconditioning, so that the subsequent response to TNF-alpha was inhibited. Our results show that sLRP-alpha is biologically active and may attenuate neuropathic pain. In the PNS, the function of LRP1 may reflect the integrated activities of the membrane-anchored and shed forms of LRP1.
Collapse
Affiliation(s)
- Alban Gaultier
- Department of Pathology, UCSD School of Medicine, La Jolla, California 92093-0629, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Montel V, Gaultier A, Lester RD, Campana WM, Gonias SL. The low-density lipoprotein receptor-related protein regulates cancer cell survival and metastasis development. Cancer Res 2007; 67:9817-24. [PMID: 17942912 DOI: 10.1158/0008-5472.can-07-0683] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Low-density lipoprotein receptor-related protein-1 (LRP-1) is a multifunctional receptor involved in receptor-mediated endocytosis and cell signaling. In this study, we show that LRP-1 is abundantly expressed in severe combined immunodeficient (SCID) mouse xenografts by various human cancer cell lines that express very low or undetectable levels of LRP-1 when cultured in 21% O2 in vitro (standard cell culture conditions). To test whether LRP-1 expression in vivo may be explained by hypoxia in the xenografts, CL16 cells, which are derived from the MDA-MB-435 cell line, were cultured in 1.0% O2. A substantial increase in LRP-1 expression was observed. To test the activity of LRP-1 in cancer progression in vivo, LRP-1 expression was silenced in CL16 cells with short hairpin RNA. These cells formed tumors in SCID mice, in which LRP-1 expression remained silenced. Although LRP-1 gene silencing did not inhibit CL16 cell dissemination from the primary tumors to the lungs, the pulmonary metastases failed to enlarge, suggesting compromised survival or growth at the implantation site. In cell culture experiments, significantly increased cell death was observed when LRP-1-silenced CL16 cells were exposed to CoCl2, which models changes that occur in hypoxia. Furthermore, LRP-1-silenced cells expressed decreased levels of vascular endothelial growth factor in response to 1.0% O2. These results suggest mechanisms by which LRP-1 may facilitate the development and growth of cancer metastases in vivo.
Collapse
Affiliation(s)
- Valérie Montel
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, California 92093-0612, USA
| | | | | | | | | |
Collapse
|
25
|
Zhang X, Polavarapu R, She H, Mao Z, Yepes M. Tissue-type plasminogen activator and the low-density lipoprotein receptor-related protein mediate cerebral ischemia-induced nuclear factor-kappaB pathway activation. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:1281-90. [PMID: 17717150 PMCID: PMC1988877 DOI: 10.2353/ajpath.2007.070472] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Tissue-type plasminogen activator (tPA) is a serine proteinase found in the intravascular space and the central nervous system. The low-density lipoprotein receptor-related protein (LRP) is a member of the low-density lipoprotein receptor gene family found in neurons and astrocytes. Cerebral ischemia induces activation of the nuclear factor (NF)-kappaB pathway. The present study investigated the role that the interaction between tPA and LRP plays on middle cerebral artery occlusion (MCAO)-induced NF-kappaB-mediated inflammatory response. We found that MCAO increased LRP expression primarily in astrocytes and that this effect was significantly decreased in the absence of tPA. The onset of the ischemic insult induced activation of the NF-kappaB pathway in wild-type and plasminogen (Plg(-/-))-deficient mice, and this effect was attenuated after inhibition of LRP or genetic deficiency of tPA. Moreover, administration of tPA to tPA(-/-) mice resulted in activation of the NF-kappaB pathway comparable with that observed in wild-type and Plg(-/-) mice. We also report that inhibition of either tPA activity or LRP or genetic deficiency of tPA resulted in a significant decrease in MCAO-induced nitric oxide production and inducible nitric-oxide synthase expression. In conclusion, our results demonstrate that after MCAO the interaction between tPA and LRP results in NF-kappaB activation in astrocytes and induction of inducible nitric-oxide synthase expression in the ischemic tissue, suggesting a cytokine-like plasminogen-independent role for tPA during cerebral ischemia.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Whitehead Biomedical Research Bldg., 615 Michael St., Suite 505J, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
26
|
Sánchez MC, Barcelona PF, Luna JD, Ortiz SG, Juarez PC, Riera CM, Chiabrando GA. Low-density lipoprotein receptor-related protein-1 (LRP-1) expression in a rat model of oxygen-induced retinal neovascularization. Exp Eye Res 2006; 83:1378-85. [PMID: 16979164 DOI: 10.1016/j.exer.2006.07.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Revised: 07/10/2006] [Accepted: 07/21/2006] [Indexed: 11/21/2022]
Abstract
The low-density lipoprotein receptor-related protein-1 (LRP-1) is a high-molecular weight receptor of the LDL receptor gene family. Its ability to bind and internalize both proteinases and proteinase-inhibitor complexes from the extracellular space suggests that it has a major role in modulating uncontrolled retinal cell proliferation. In order to test this assumption, we investigated the expression of LRP-1 and receptor-associated ligands in a rat model of oxygen-induced retinal neovascularization. Wistar albino rats were placed into incubators at birth and exposed to an atmosphere alternating between 50% and 10% of oxygen every 24 h. After 14 days, the animals were allowed to recover in room air and sacrificed at postnatal day 20 (P20). The protein expression of LRP-1 and alpha2-macroglobulin (alpha2M) in the retina from unexposed and hyperoxia-exposed rats was investigated by Western blot. The localization of LRP-1 after neovascularization was assessed by immunohistochemical staining. The activity of metalloproteinases (MMPs) was determined by zymography. Histological analysis was done to quantitate the neovascular response in these animals. Western blot analysis showed that LRP-1 was expressed, along with alpha2M, in the retina of rats with oxygen-induced neovascularization at P20. By immunohistochemical analysis, positive staining for LRP-1 appeared in cells extending from the inner limiting membrane (ILM) to the outer limiting membrane (OLM). The cells of the retina that expressed LRP-1 were identified by immunofluorescence as Müller cells. Zymographic analysis demonstrated increased activity of MMP-2 and MMP-9 under neovascular conditions. This is the first demonstration of the involvement of LRP-1 in retinal neovascularization. In retinas of rats with oxygen-induced neovascularization, the expression of LRP-1 and alpha2M was increased along with an enhanced activity of MMPs, suggesting that LRP-1 expression may play a role in modulating retinal neovascularization by regulating proteolytic activity.
Collapse
Affiliation(s)
- María C Sánchez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria (5000), Córdoba, Argentina.
| | | | | | | | | | | | | |
Collapse
|
27
|
Campana WM, Li X, Dragojlovic N, Janes J, Gaultier A, Gonias SL. The low-density lipoprotein receptor-related protein is a pro-survival receptor in Schwann cells: possible implications in peripheral nerve injury. J Neurosci 2006; 26:11197-207. [PMID: 17065459 PMCID: PMC6674644 DOI: 10.1523/jneurosci.2709-06.2006] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Schwann cells undergo phenotypic modulation in peripheral nerve injury. In the adult rodent, Schwann cells are resistant to death-promoting challenges. The responsible receptors and signaling pathways are incompletely understood. In this study, we demonstrate that low-density lipoprotein receptor-related protein-1 (LRP-1) is expressed in adult sciatic nerve. After crush injury, LRP-1 is lost from the axoplasm and substantially upregulated in Schwann cells. Increased LRP-1 mRNA expression was observed locally at the injury site in multiple forms of sciatic nerve injury, including crush injury, chronic constriction injury, and axotomy. Endogenously produced tumor necrosis factor-alpha (TNF-alpha) was mostly responsible for the increase in LRP-1 expression; this activity was reproduced by direct injection of TNF-alpha into injured nerves in the TNF-alpha gene knock-out mouse. TNF receptor II was primarily involved. TNF-alpha also increased LRP-1 mRNA in Schwann cells in primary culture. Silencing of Schwann cell LRP-1 with siRNA decreased phosphorylated Akt and increased activated caspase-3. Equivalent changes in cell signaling were observed in LRP-1-deficient murine embryonic fibroblasts. Schwann cell death was induced in vitro by serum withdrawal or TNF-alpha, to a greater extent when LRP-1 was silenced. Schwann cell death was induced in vivo by injecting the LRP-1 antagonist, receptor-associated protein, into axotomy sites in adult rats. These results support a model in which LRP-1 functions as a pro-survival receptor in Schwann cells.
Collapse
Affiliation(s)
- W Marie Campana
- Department of Anesthesiology, University of California, San Diego School of Medicine, La Jolla, California 92093-0629, USA.
| | | | | | | | | | | |
Collapse
|