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
|
David T, du Roure PD, Mallavialle A, Laurent-Matha V, Roger P, Guiu S, Chardès T, Liaudet-Coopman E. Cathepsins: Novel opportunities for antibody therapeutics in cancer. Br J Pharmacol 2025; 182:1671-1682. [PMID: 39834229 DOI: 10.1111/bph.17437] [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: 08/08/2024] [Revised: 12/06/2024] [Accepted: 12/07/2024] [Indexed: 01/22/2025] Open
Abstract
Cathepsins, the most abundant lysosomal proteases, have key functions in cell maintenance and homeostasis. They are overexpressed and hypersecreted in cancer and associated with poor prognosis. Secreted cathepsins display pro-tumour activities in the tumour microenvironment and thus represent interesting molecular targets in oncology. Recently, several antibody-based cancer therapies have targeted the pro-tumour activity of the extracellular cathepsin pool, altering several cancer hallmarks, but not the intracellular cathepsin levels that are often crucial for cell homeostasis. In this mini-review, we describe advances in antibodies against extracellular cathepsins in cancer, and their effect on the proteolytic cascade, matrix remodelling, proliferation, and modulation of the anti-cancer immune response. We also discuss the add-on value of combination strategies (anti-cathepsin antibodies with chemotherapy and/or biologics) that make anti-cathepsin antibodies a new opportunity for disease management.
Collapse
Affiliation(s)
- Timothée David
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Aude Mallavialle
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Pascal Roger
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- Department of Pathology, CHU Nîmes, Nîmes, France
| | - Séverine Guiu
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- Department of Medical Oncology, ICM, Montpellier, France
| | - Thierry Chardès
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- Centre National de la Recherche Scientifique, CNRS, Paris, France
| | | |
Collapse
|
3
|
Niu L, Zhou X, Li D, Zheng Y, Li H. Glycosylation Triggers Cathepsin D Maturation and Secretion to Promote Gastric Cancer Development. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00081-1. [PMID: 40122458 DOI: 10.1016/j.ajpath.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/20/2025] [Accepted: 02/28/2025] [Indexed: 03/25/2025]
Abstract
Cathepsin D (CTSD) is a lysosomal aspartic protease with high expression in cancers. CTSD localized in different subcellular regions performs distinct roles. However, the precise regulation of its intracellular trafficking and extracellular secretion remains incompletely understood. This study showed that glycosylation modifications of CTSD determine its maturation and secretion in gastric cancer (GC) cells. Specifically, glycosylation at asparagine 134 (N134) dictated the intracellular trafficking and maturation of CTSD within lysosomes, through facilitating its sorting into COPII vesicles. Glycosylation at asparagine 263 (N263) was essential for secretion of the proenzyme form of CTSD (pro-CTSD) via a novel pathway dependent on the small GTPase Rab3D. Notably, the extracellular release of pro-CTSD occurred more rapidly than its intracellular trafficking from the endoplasmic reticulum to lysosomes. This enhanced secretion speed may rapidly elevate the levels of pro-CTSD in the tumor microenvironment in response to extracellular stimuli. Ultimately, glycosylation at N134 and N263 regulated the autophagy and cell proliferation, respectively. These findings show the role of glycosylation in triggering the maturation and secretion of CTSD in GC cells. Through modulating its cellular trafficking, differential glycosylation modifications of CTSD defined the malignant behavior of GC cells.
Collapse
Affiliation(s)
- Liling Niu
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China; Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Xunzhu Zhou
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China; Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Deman Li
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China; Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Yongye Zheng
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China; Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Hui Li
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China; Tianjin Key Laboratory of Digestive Cancer, Tianjin, China.
| |
Collapse
|
4
|
Wan M, Pan S, Shan B, Diao H, Jin H, Wang Z, Wang W, Han S, Liu W, He J, Zheng Z, Pan Y, Han X, Zhang J. Lipid metabolic reprograming: the unsung hero in breast cancer progression and tumor microenvironment. Mol Cancer 2025; 24:61. [PMID: 40025508 PMCID: PMC11874147 DOI: 10.1186/s12943-025-02258-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/02/2025] [Indexed: 03/04/2025] Open
Abstract
Aberrant lipid metabolism is a well-recognized hallmark of cancer. Notably, breast cancer (BC) arises from a lipid-rich microenvironment and depends significantly on lipid metabolic reprogramming to fulfill its developmental requirements. In this review, we revisit the pivotal role of lipid metabolism in BC, underscoring its impact on the progression and tumor microenvironment. Firstly, we delineate the overall landscape of lipid metabolism in BC, highlighting its roles in tumor progression and patient prognosis. Given that lipids can also act as signaling molecules, we next describe the lipid signaling exchanges between BC cells and other cellular components in the tumor microenvironment. Additionally, we summarize the therapeutic potential of targeting lipid metabolism from the aspects of lipid metabolism processes, lipid-related transcription factors and immunotherapy in BC. Finally, we discuss the possibilities and problems associated with clinical applications of lipid‑targeted therapy in BC, and propose new research directions with advances in spatiotemporal multi-omics.
Collapse
Affiliation(s)
- Mengting Wan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuaikang Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Benjie Shan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Haizhou Diao
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwei Jin
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Ziqi Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Shuya Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wan Liu
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiaying He
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Graduate School of Bengbu Medical University, Bengbu, Anhui Province, China
| | - Zihan Zheng
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Yueyin Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Jinguo Zhang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
5
|
Desroys du Roure P, David T, Mallavialle A, Laurent-Matha V, Roger P, Guiu S, Chardès T, Liaudet-Coopman E. Antibodies against the multifaceted cathepsin D protein open new avenues for TNBC immunotherapy. J Immunother Cancer 2025; 13:e009548. [PMID: 39800383 PMCID: PMC11748927 DOI: 10.1136/jitc-2024-009548] [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: 04/26/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous breast cancer subtype characterized by aggressive clinical behavior and poor prognosis. The immune landscape associated with TNBC often reveals high immunogenicity. Therefore, immunotherapy, which has demonstrated its efficacy in different cancer types, could be a promising strategy for TNBC, given the limited therapeutic options currently available besides conventional chemotherapy. The aspartic protease cathepsin D (cath-D) is a tumor cell-associated extracellular protein with protumor activity, a marker of poor prognosis, and a target for antibody-based therapy in TNBC. This commentary provides a synopsis/narrative summary of the development of anti-cath-D antibodies in different formats, their key roles in restoring the antitumor immunity, particularly via activation of tumor-infiltrating natural killer cells, and their dual antitumor effects on cancer cells and stromal cancer-associated fibroblasts, suggesting their interest for clinical use in the light of the current clinical knowledge on TNBC.
Collapse
Affiliation(s)
| | - Timothée David
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Aude Mallavialle
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Pascal Roger
- Department of Pathology, CHU Nîmes, Nîmes, France
| | - Séverine Guiu
- Department of Medical Oncology, ICM, Montpellier, France
| | - Thierry Chardès
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- CNRS, Centre National de la Recherche Scientifique, Paris, F-75016, France
| | | |
Collapse
|
6
|
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
|
7
|
Zhao D, Huang ZK, Liang Y, Li ZJ, Zhang XW, Li KH, Wu H, Zhang XD, Li CS, An D, Sun X, An MX, Shi JX, Bao YJ, Tian L, Wang DF, Wu AH, Chen YH, Zhao WD. Monocytes Release Pro-Cathepsin D to Drive Blood-to-Brain Transcytosis in Diabetes. Circ Res 2024; 134:e17-e33. [PMID: 38420756 DOI: 10.1161/circresaha.123.323622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Microvascular complications are the major outcome of type 2 diabetes progression, and the underlying mechanism remains to be determined. METHODS High-throughput RNA sequencing was performed using human monocyte samples from controls and diabetes. The transgenic mice expressing human CTSD (cathepsin D) in the monocytes was constructed using CD68 promoter. In vivo 2-photon imaging, behavioral tests, immunofluorescence, transmission electron microscopy, Western blot analysis, vascular leakage assay, and single-cell RNA sequencing were performed to clarify the phenotype and elucidate the molecular mechanism. RESULTS Monocytes expressed high-level CTSD in patients with type 2 diabetes. The transgenic mice expressing human CTSD in the monocytes showed increased brain microvascular permeability resembling the diabetic microvascular phenotype, accompanied by cognitive deficit. Mechanistically, the monocytes release nonenzymatic pro-CTSD to upregulate caveolin expression in brain endothelium triggering caveolae-mediated transcytosis, without affecting the paracellular route of brain microvasculature. The circulating pro-CTSD activated the caveolae-mediated transcytosis in brain endothelial cells via its binding with low-density LRP1 (lipoprotein receptor-related protein 1). Importantly, genetic ablation of CTSD in the monocytes exhibited a protective effect against the diabetes-enhanced brain microvascular transcytosis and the diabetes-induced cognitive impairment. CONCLUSIONS These findings uncover the novel role of circulatory pro-CTSD from monocytes in the pathogenesis of cerebral microvascular lesions in diabetes. The circulatory pro-CTSD is a potential target for the intervention of microvascular complications in diabetes.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Zeng-Kang Huang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Yu Liang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Zhi-Jun Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Xue-Wei Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Kun-Hang Li
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Hao Wu
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Xu-Dong Zhang
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Chen-Sheng Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Dong An
- School of Mechanical Engineering, Shenyang Jianzhu University, China (D.A.)
| | - Xue Sun
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Ming-Xin An
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Jun-Xiu Shi
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Yi-Jun Bao
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Li Tian
- Department of Gerontology (L.T., D.-F.W.), Shengjing Hospital of China Medical University, Shenyang, China
| | - Di-Fei Wang
- Department of Gerontology (L.T., D.-F.W.), Shengjing Hospital of China Medical University, Shenyang, China
| | - An-Hua Wu
- Department of Neurosurgery (A.-H.W.), Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu-Hua Chen
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Wei-Dong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| |
Collapse
|
8
|
Desroys du Roure P, Lajoie L, Mallavialle A, Alcaraz LB, Mansouri H, Fenou L, Garambois V, Rubio L, David T, Coenon L, Boissière-Michot F, Chateau MC, Ngo G, Jarlier M, Villalba M, Martineau P, Laurent-Matha V, Roger P, Guiu S, Chardès T, Gros L, Liaudet-Coopman E. A novel Fc-engineered cathepsin D-targeting antibody enhances ADCC, triggers tumor-infiltrating NK cell recruitment, and improves treatment with paclitaxel and enzalutamide in triple-negative breast cancer. J Immunother Cancer 2024; 12:e007135. [PMID: 38290768 PMCID: PMC10828871 DOI: 10.1136/jitc-2023-007135] [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] [Accepted: 12/19/2023] [Indexed: 02/01/2024] Open
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) prognosis is poor. Immunotherapies to enhance the antibody-induced natural killer (NK) cell antitumor activity are emerging for TNBC that is frequently immunogenic. The aspartic protease cathepsin D (cath-D), a tumor cell-associated extracellular protein with protumor activity and a poor prognosis marker in TNBC, is a prime target for antibody-based therapy to induce NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC). This study investigated whether Fc-engineered anti-cath-D antibodies trigger ADCC, their impact on antitumor efficacy and tumor-infiltrating NK cells, and their relevance for combinatory therapy in TNBC. METHODS Cath-D expression and localization in TNBC samples were evaluated by western blotting, immunofluorescence, and immunohistochemistry. The binding of human anti-cath-D F1M1 and Fc-engineered antibody variants, which enhance (F1M1-Fc+) or prevent (F1M1-Fc-) affinity for CD16a, to secreted human and murine cath-D was analyzed by ELISA, and to CD16a by surface plasmon resonance and flow cytometry. NK cell activation was investigated by flow cytometry, and ADCC by lactate dehydrogenase release. The antitumor efficacy of F1M1 Fc-variants was investigated using TNBC cell xenografts in nude mice. NK cell recruitment, activation, and cytotoxic activity were analyzed in MDA-MB-231 cell xenografts by immunophenotyping and RT-qPCR. NK cells were depleted using an anti-asialo GM1 antibody. F1M1-Fc+ antitumor effect was assessed in TNBC patient-derived xenografts (PDXs) and TNBC SUM159 cell xenografts, and in combination with paclitaxel or enzalutamide. RESULTS Cath-D expression on the TNBC cell surface could be exploited to induce ADCC. F1M1 Fc-variants recognized human and mouse cath-D. F1M1-Fc+ activated NK cells in vitro and induced ADCC against TNBC cells and cancer-associated fibroblasts more efficiently than F1M1. F1M1-Fc- was ineffective. In the MDA-MB-231 cell xenograft model, F1M1-Fc+ displayed higher antitumor activity than F1M1, whereas F1M1-Fc- was less effective, reflecting the importance of Fc-dependent mechanisms in vivo. F1M1-Fc+ triggered tumor-infiltrating NK cell recruitment, activation and cytotoxic activity in MDA-MB-231 cell xenografts. NK cell depletion impaired F1M1-Fc+ antitumor activity, demonstrating their key role. F1M1-Fc+ inhibited growth of SUM159 cell xenografts and two TNBC PDXs. In combination therapy, F1M1-Fc+ improved paclitaxel and enzalutamide therapeutic efficacy without toxicity. CONCLUSIONS F1M1-Fc+ is a promising immunotherapy for TNBC that could be combined with conventional regimens, including chemotherapy or antiandrogens.
Collapse
Affiliation(s)
| | - Laurie Lajoie
- Université de Tours - INRAE, UMR1282, Infectiologie et Santé Publique (ISP), équipe BioMédicaments Anti-Parasitaires (BioMAP), Tours, France
| | - Aude Mallavialle
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Lindsay B Alcaraz
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Hanane Mansouri
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- RHEM, IRCM, Montpellier, France
| | - Lise Fenou
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Lucie Rubio
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Timothée David
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Loïs Coenon
- IRMB, University of Montpellier, INSERM, CNRS, CHU Montpellier, Montpellier, France
| | | | | | - Giang Ngo
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Martin Villalba
- IRMB, University of Montpellier, INSERM, CNRS, CHU Montpellier, Montpellier, France
- Institut du Cancer Avignon-Provence Sainte Catherine, Avignon, France
| | - Pierre Martineau
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Pascal Roger
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- Department of Pathology, CHU Nîmes, Nimes, France
| | - Séverine Guiu
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- Department of Medical Oncology, ICM, Montpellier, France
| | - Thierry Chardès
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- CNRS, Centre national de la recherche Scientifique, Paris, F-75016, France
| | - Laurent Gros
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- CNRS, Centre national de la recherche Scientifique, Paris, F-75016, France
| | | |
Collapse
|
9
|
David T, Mallavialle A, Faget J, Alcaraz LB, Lapierre M, du Roure PD, Laurent-Matha V, Mansouri H, Jarlier M, Martineau P, Roger P, Guiu S, Chardès T, Liaudet-Coopman E. Anti-cathepsin D immunotherapy triggers both innate and adaptive anti-tumour immunity in breast cancer. Br J Pharmacol 2023. [PMID: 38030588 DOI: 10.1111/bph.16291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Triple-negative breast cancer (TNBC) has poorer outcomes than other breast cancers (BC), including HER2+ BC. Cathepsin D (CathD) is a poor prognosis marker overproduced by BC cells, hypersecreted in the tumour microenvironment with tumour-promoting activity. Here, we characterized the immunomodulatory activity of the anti-CathD antibody F1 and its improved Fab-aglycosylated version (F1M1) in immunocompetent mouse models of TNBC (C57BL/6 mice harbouring E0771 cell grafts) and HER2-amplified BC (BALB/c mice harbouring TUBO cell grafts). EXPERIMENTAL APPROACH CathD expression was evaluated by western blotting and immunofluorescence, and antibody binding to CathD by ELISA. Antibody anti-tumour efficacy was investigated in mouse models. Immune cell recruitment and activation were assessed by immunohistochemistry, immunophenotyping, and RT-qPCR. KEY RESULTS F1 and F1M1 antibodies remodelled the tumour immune landscape. Both antibodies promoted innate antitumour immunity by preventing the recruitment of immunosuppressive M2-polarized tumour-associated macrophages (TAMs) and by activating natural killer cells in the tumour microenvironment of both models. This translated into a reduction of T-cell exhaustion markers in the tumour microenvironment that could be locally supported by enhanced activation of anti-tumour antigen-presenting cell (M1-polarized TAMs and cDC1 cells) functions. Both antibodies inhibited tumour growth in the highly-immunogenic E0771 model, but only marginally in the immune-excluded TUBO model, indicating that anti-CathD immunotherapy is more relevant for BC with a high immune cell infiltrate, as often observed in TNBC. CONCLUSION AND IMPLICATION Anti-CathD antibody-based therapy triggers the anti-tumour innate and adaptive immunity in preclinical models of BC and is a promising immunotherapy for immunogenic TNBC.
Collapse
Affiliation(s)
- Timothée David
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
| | | | - Julien Faget
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
| | | | - Marion Lapierre
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
| | | | | | - Hanane Mansouri
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
- RHEM, IRCM, Montpellier, France
| | | | | | - Pascal Roger
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
- Department of Pathology, CHU Nîmes, Nîmes, France
| | - Séverine Guiu
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
- Department of Medical Oncology, ICM, Montpellier, France
| | - Thierry Chardès
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
- Centre national de la recherche Scientifique, CNRS, Paris, France
| | | |
Collapse
|
10
|
Liu Y, Jian J, Zhang Y, Wang L, Liu X, Chen Z. Construction of cancer- associated fibroblasts related risk signature based on single-cell RNA-seq and bulk RNA-seq data in bladder urothelial carcinoma. Front Oncol 2023; 13:1170893. [PMID: 37124542 PMCID: PMC10140328 DOI: 10.3389/fonc.2023.1170893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Background The ability of cancer-associated fibroblasts (CAFs) to encourage angiogenesis, tumor cell spread, and increase treatment resistance makes them pro-tumorigenic. We aimed to investigate the CAF signature in Bladder urothelial carcinoma (BLCA) and, for clinical application, to build a CAF-based risk signature to decipher the immune landscape and screen for suitable treatment BLCA samples. Methods CAF-related genes were discovered by superimposing CAF marker genes discovered from single-cell RNA-seq (scRNA-seq) data taken from the GEO database with CAF module genes discovered by weighted gene co-expression network analysis (WGCNA) using bulk RNA-seq data from TCGA. After identifying prognostic genes related with CAF using univariate Cox regression, Lasso regression was used to build a risk signature. With microarray data from the GEO database, prognostic characteristics were externally verified. For high and low CAF-risk categories, immune cells and immunotherapy responses were analyzed. Finally, a nomogram model based on the risk signature and prospective chemotherapeutic drugs were examined. Results Combining scRNA-seq and bulk-seq data analysis yielded a total of 124 CAF-related genes. LRP1, ANXA5, SERPINE2, ECM1, RBP1, GJA1, and FKBP10 were the seven BLCA prognostic genes that remained after univariate Cox regression and LASSO regression analyses. Then, based on these genes, prognostic characteristics were created and validated to predict survival in BLCA patients. Additionally, risk signature had a strong correlation with known CAF scores, stromal scores, and certain immune cells. The CAF-risk signature was identified as an independent prognostic factor for BLCA using multifactorial analysis, and its usefulness in predicting immunotherapy response was confirmed. Based on risk classification, we projected six highly sensitive anticancer medicines for the high-risk group. Conclusion The prognosis of BLCA may be accurately predicted using CAF-based risk signature. With a thorough understanding of the BLCA CAF-signature, it might be able to explain the BLCA patients' response to immunotherapy and identify a potential target for BLCA treatment.
Collapse
Affiliation(s)
- Yunxun Liu
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, China
| | - Jun Jian
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, China
| | - Ye Zhang
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, China
| | - Lei Wang
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, China
- *Correspondence: Xiuheng Liu, ; Zhiyuan Chen,
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, China
- *Correspondence: Xiuheng Liu, ; Zhiyuan Chen,
| |
Collapse
|
11
|
Yamamoto K, Scavenius C, Meschis MM, Gremida AME, Mogensen EH, Thøgersen IB, Bonelli S, Scilabra SD, Jensen A, Santamaria S, Ahnström J, Bou-Gharios G, Enghild JJ, Nagase H. A top-down approach to uncover the hidden ligandome of low-density lipoprotein receptor-related protein 1 in cartilage. Matrix Biol 2022; 112:190-218. [PMID: 36028175 DOI: 10.1016/j.matbio.2022.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/26/2022] [Accepted: 08/17/2022] [Indexed: 11/29/2022]
Abstract
The low-density lipoprotein receptor-related protein 1 (LRP1) is a cell-surface receptor ubiquitously expressed in various tissues. It plays tissue-specific roles by mediating endocytosis of a diverse range of extracellular molecules. Dysregulation of LRP1 is involved in multiple conditions including osteoarthritis (OA) but little information is available about the specific profile of direct binding partners of LRP1 (ligandome) for each tissue, which would lead to a better understanding of its role in disease states. Here, we investigated adult articular cartilage where impaired LRP1-mediated endocytosis leads to tissue destruction. We used a top-down approach involving proteomic analysis of the LRP1 interactome in human chondrocytes, direct binding assays using purified LRP1 and ligand candidates, and validation in LRP1-deficient fibroblasts and human chondrocytes, as well as a novel Lrp1 conditional knockout (KO) mouse model. We found that inhibition of LRP1 and ligand interaction results in cell death, alteration of the entire secretome and transcriptional modulations in human chondrocytes. We identified a chondrocyte-specific LRP1 ligandome consisting of more than 50 novel ligand candidates. Surprisingly, 23 previously reported LRP1 ligands were not regulated by LRP1-mediated endocytosis in human chondrocytes. We confirmed direct LRP1 binding of HGFAC, HMGB1, HMGB2, CEMIP, SLIT2, ADAMTS1, TSG6, IGFBP7, SPARC and LIF, correlation between their affinity for LRP1 and the rate of endocytosis, and some of their intracellular localization. Moreover, a conditional LRP1 KO mouse model demonstrated a critical role of LRP1 in regulating the high-affinity ligands in cartilage in vivo. This systematic approach revealed the specificity and the extent of the chondrocyte LRP1 ligandome and identified potential novel therapeutic targets for OA.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom.
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Maria M Meschis
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Abdulrahman M E Gremida
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Emilie H Mogensen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Ida B Thøgersen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Simone Bonelli
- Fondazione RiMED - ISMETT via Ernesto Tricomi 5, 90127 Palermo, Italy
| | - Simone D Scilabra
- Fondazione RiMED - ISMETT via Ernesto Tricomi 5, 90127 Palermo, Italy
| | - Anders Jensen
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Salvatore Santamaria
- Department of Immunology and Inflammation, Imperial College London, Du Cane Road, W12 0NN, London, United Kingdom
| | - Josefin Ahnström
- Department of Immunology and Inflammation, Imperial College London, Du Cane Road, W12 0NN, London, United Kingdom
| | - George Bou-Gharios
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Hideaki Nagase
- Kennedy Institute of Rheumatology, University of Oxford, Headington, Oxford OX3 7FY, United Kingdom
| |
Collapse
|
12
|
Alcaraz LB, Mallavialle A, David T, Derocq D, Delolme F, Dieryckx C, Mollevi C, Boissière-Michot F, Simony-Lafontaine J, Du Manoir S, Huesgen PF, Overall CM, Tartare-Deckert S, Jacot W, Chardès T, Guiu S, Roger P, Reinheckel T, Moali C, Liaudet-Coopman E. A 9-kDa matricellular SPARC fragment released by cathepsin D exhibits pro-tumor activity in the triple-negative breast cancer microenvironment. Am J Cancer Res 2021; 11:6173-6192. [PMID: 33995652 PMCID: PMC8120228 DOI: 10.7150/thno.58254] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/20/2021] [Indexed: 12/26/2022] Open
Abstract
Rationale: Alternative therapeutic strategies based on tumor-specific molecular targets are urgently needed for triple-negative breast cancer (TNBC). The protease cathepsin D (cath-D) is a marker of poor prognosis in TNBC and a tumor-specific extracellular target for antibody-based therapy. The identification of cath-D substrates is crucial for the mechanistic understanding of its role in the TNBC microenvironment and future therapeutic developments. Methods: The cath-D substrate repertoire was investigated by N-Terminal Amine Isotopic Labeling of Substrates (TAILS)-based degradome analysis in a co-culture assay of TNBC cells and breast fibroblasts. Substrates were validated by amino-terminal oriented mass spectrometry of substrates (ATOMS). Cath-D and SPARC expression in TNBC was examined using an online transcriptomic survival analysis, tissue micro-arrays, TNBC cell lines, patient-derived xenografts (PDX), human TNBC samples, and mammary tumors from MMTV-PyMT Ctsd-/-knock-out mice. The biological role of SPARC and its fragments in TNBC were studied using immunohistochemistry and immunofluorescence analysis, gene expression knockdown, co-culture assays, western blot analysis, RT-quantitative PCR, adhesion assays, Transwell motility, trans-endothelial migration and invasion assays. Results: TAILS analysis showed that the matricellular protein SPARC is a substrate of extracellular cath-D. In vitro, cath-D induced limited proteolysis of SPARC C-terminal extracellular Ca2+ binding domain at acidic pH, leading to the production of SPARC fragments (34-, 27-, 16-, 9-, and 6-kDa). Similarly, cath-D secreted by TNBC cells cleaved fibroblast- and cancer cell-derived SPARC at the tumor pericellular acidic pH. SPARC cleavage also occurred in TNBC tumors. Among these fragments, only the 9-kDa SPARC fragment inhibited TNBC cell adhesion and spreading on fibronectin, and stimulated their migration, endothelial transmigration, and invasion. Conclusions: Our study establishes a novel crosstalk between proteases and matricellular proteins in the tumor microenvironment through limited SPARC proteolysis, revealing a novel targetable 9-kDa bioactive SPARC fragment for new TNBC treatments. Our study will pave the way for the development of strategies for targeting bioactive fragments from matricellular proteins in TNBC.
Collapse
|
13
|
Abstract
Epidemiological studies have reported an inverse correlation between cancer and neurodegenerative disorders, and increasing evidence shows that similar genes and pathways are dysregulated in both diseases but in a contrasting manner. Given the genetic convergence of the neuronal ceroid lipofuscinoses (NCLs), a family of rare neurodegenerative disorders commonly known as Batten disease, and other neurodegenerative diseases, we sought to explore the relationship between cancer and the NCLs. In this review, we survey data from The Cancer Genome Atlas and available literature on the roles of NCL genes in different oncogenic processes to reveal links between all the NCL genes and cancer-related processes. We also discuss the potential contributions of NCL genes to cancer immunology. Based on our findings, we propose that further research on the relationship between cancer and the NCLs may help shed light on the roles of NCL genes in both diseases and possibly guide therapy development.
Collapse
|
14
|
Goyal S, Patel KV, Nagare Y, Raykar DB, Raikar SS, Dolas A, Khurana P, Cyriac R, Sarak S, Gangar M, Agarwal AK, Kulkarni A. Identification and structure-activity relationship studies of small molecule inhibitors of the human cathepsin D. Bioorg Med Chem 2020; 29:115879. [PMID: 33271453 DOI: 10.1016/j.bmc.2020.115879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/08/2020] [Accepted: 11/13/2020] [Indexed: 01/18/2023]
Abstract
Cathepsin D, an aspartyl protease, is an attractive therapeutic target for various diseases, primarily cancer and osteoarthritis. However, despite several small molecule cathepsin D inhibitors being developed, that are highly potent, most of them show poor microsomal stability, which in turn limits their clinical translation. Herein, we describe the design, optimization and evaluation of a series of novel non-peptidic acylguanidine based small molecule inhibitors of cathepsin D. Optimization of our hit compound 1a (IC50 = 29 nM) led to the highly potent mono sulphonamide analogue 4b (IC50 = 4 nM), however with poor microsomal stability (HLM: 177 and MLM: 177 μl/min/mg). To further improve the microsomal stability while retaining the potency, we carried out an extensive structure-activity relationship screen which led to the identification of our optimised lead 24e (IC50 = 45 nM), with an improved microsomal stability (HLM: 59.1 and MLM: 86.8 μl/min/mg). Our efforts reveal that 24e could be a good starting point or potential candidate for further preclinical studies against diseases where Cathepsin D plays an important role.
Collapse
Affiliation(s)
| | | | - Yadav Nagare
- Aten Porus Lifesciences, Bangalore 560068, India
| | | | | | - Atul Dolas
- Aten Porus Lifesciences, Bangalore 560068, India
| | | | | | - Sharad Sarak
- Aten Porus Lifesciences, Bangalore 560068, India
| | | | - Anil K Agarwal
- Department of Chemistry, CHRIST (Deemed to be University), Bengaluru, Karnataka, India
| | - Aditya Kulkarni
- Aten Porus Lifesciences, Bangalore 560068, India; Avaliv Therapeutics, Naples, FL, USA.
| |
Collapse
|
15
|
Smith TA, Ghergherehchi CL, Tucker HO, Bittner GD. Coding transcriptome analyses reveal altered functions underlying immunotolerance of PEG-fused rat sciatic nerve allografts. J Neuroinflammation 2020; 17:287. [PMID: 33008419 PMCID: PMC7532577 DOI: 10.1186/s12974-020-01953-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 09/16/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Current methods to repair ablation-type peripheral nerve injuries (PNIs) using peripheral nerve allografts (PNAs) often result in poor functional recovery due to immunological rejection as well as to slow and inaccurate outgrowth of regenerating axonal sprouts. In contrast, ablation-type PNIs repaired by PNAs, using a multistep protocol in which one step employs the membrane fusogen polyethylene glycol (PEG), permanently restore sciatic-mediated behaviors within weeks. Axons and cells within PEG-fused PNAs remain viable, even though outbred host and donor tissues are neither immunosuppressed nor tissue matched. PEG-fused PNAs exhibit significantly reduced T cell and macrophage infiltration, expression of major histocompatibility complex I/II and consistently low apoptosis. In this study, we analyzed the coding transcriptome of PEG-fused PNAs to examine possible mechanisms underlying immunosuppression. METHODS Ablation-type sciatic PNIs in adult Sprague-Dawley rats were repaired using PNAs and a PEG-fusion protocol combined with neurorrhaphy. Electrophysiological and behavioral tests confirmed successful PEG-fusion of PNAs. RNA sequencing analyzed differential expression profiles of protein-coding genes between PEG-fused PNAs and negative control PNAs (not treated with PEG) at 14 days PO, along with unoperated control nerves. Sequencing results were validated by quantitative reverse transcription PCR (RT-qPCR), and in some cases, immunohistochemistry. RESULTS PEG-fused PNAs display significant downregulation of many gene transcripts associated with innate and adaptive allorejection responses. Schwann cell-associated transcripts are often upregulated, and cellular processes such as extracellular matrix remodeling and cell/tissue development are particularly enriched. Transcripts encoding several potentially immunosuppressive proteins (e.g., thrombospondins 1 and 2) also are upregulated in PEG-fused PNAs. CONCLUSIONS This study is the first to characterize the coding transcriptome of PEG-fused PNAs and to identify possible links between alterations of the extracellular matrix and suppression of the allorejection response. The results establish an initial molecular basis to understand mechanisms underlying PEG-mediated immunosuppression.
Collapse
Affiliation(s)
- Tyler A Smith
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, 78712, USA
| | | | - Haley O Tucker
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, 78712, USA
| | - George D Bittner
- Department of Neuroscience, University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
16
|
Le CC, Bennasroune A, Collin G, Hachet C, Lehrter V, Rioult D, Dedieu S, Morjani H, Appert-Collin A. LRP-1 Promotes Colon Cancer Cell Proliferation in 3D Collagen Matrices by Mediating DDR1 Endocytosis. Front Cell Dev Biol 2020; 8:412. [PMID: 32582700 PMCID: PMC7283560 DOI: 10.3389/fcell.2020.00412] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/04/2020] [Indexed: 12/19/2022] Open
Abstract
Low density lipoprotein receptor related protein-1 (LRP-1) is a large ubiquitous endocytic receptor mediating the clearance of various molecules from the extracellular matrix. Several studies have shown that LRP-1 plays crucial roles during tumorigenesis functioning as a main signal pathway regulator, especially by interacting with other cell-surface receptors. Discoïdin Domain Receptors (DDRs), type I collagen receptors with tyrosine kinase activity, have previously been associated with tumor invasion and aggressiveness in diverse tumor environments. Here, we addressed whether it could exist functional interplays between LRP-1 and DDR1 to control colon carcinoma cell behavior in three-dimensional (3D) collagen matrices. We found that LRP-1 established tight molecular connections with DDR1 at the plasma membrane in colon cancer cells. In this tumor context, we provide evidence that LRP-1 regulates by endocytosis the cell surface levels of DDR1 expression. The LRP-1 mediated endocytosis of DDR1 increased cell proliferation by promoting cell cycle progression into S phase and decreasing apoptosis. In this study, we identified a new molecular way that controls the cell-surface expression of DDR1 and consequently the colon carcinoma cell proliferation and apoptosis and highlighted an additional mechanism by which LRP-1 carries out its sensor activity of the tumor microenvironment.
Collapse
Affiliation(s)
- Cao Cuong Le
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France.,Unité BioSpecT, EA7506, Reims, France
| | - Amar Bennasroune
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Guillaume Collin
- Université de Reims Champagne-Ardenne, Reims, France.,Unité BioSpecT, EA7506, Reims, France
| | - Cathy Hachet
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Véronique Lehrter
- Université de Reims Champagne-Ardenne, Reims, France.,Unité BioSpecT, EA7506, Reims, France
| | - Damien Rioult
- Plateau Technique Mobile de Cytométrie Environnementale MOBICYTE, URCA/INERIS, Reims Champagne-Ardenne University (URCA), Reims, France
| | - Stéphane Dedieu
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Hamid Morjani
- Université de Reims Champagne-Ardenne, Reims, France.,Unité BioSpecT, EA7506, Reims, France
| | - Aline Appert-Collin
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| |
Collapse
|
17
|
Mansouri H, Alcaraz LB, Mollevi C, Mallavialle A, Jacot W, Boissière-Michot F, Simony-Lafontaine J, Laurent-Matha V, Roger P, Liaudet-Coopman E, Guiu S. Co-Expression of Androgen Receptor and Cathepsin D Defines a Triple-Negative Breast Cancer Subgroup with Poorer Overall Survival. Cancers (Basel) 2020; 12:cancers12051244. [PMID: 32429078 PMCID: PMC7281089 DOI: 10.3390/cancers12051244] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 01/21/2023] Open
Abstract
Background: In the triple-negative breast cancer (TNBC) group, the luminal androgen receptor subtype is characterized by expression of androgen receptor (AR) and lack of estrogen receptor and cytokeratin 5/6 expression. Cathepsin D (Cath-D) is overproduced and hypersecreted by breast cancer (BC) cells and is a poor prognostic marker. We recently showed that in TNBC, Cath-D is a potential target for antibody-based therapy. This study evaluated the frequency of AR/Cath-D co-expression and its prognostic value in a large series of patients with non-metastatic TNBC. Methods: AR and Cath-D expression was evaluated by immunohistochemistry in 147 non-metastatic TNBC. The threshold for AR positivity (AR+) was set at ≥1% of stained cells, and the threshold for Cath-D positivity (Cath-D+) was moderate/strong staining intensity. Lymphocyte density, macrophage infiltration, PD-L1 and programmed cell death (PD-1) expression were assessed. Results: Scarff-Bloom-Richardson grade 1–2 and lymph node invasion were more frequent, while macrophage infiltration was less frequent in AR+/Cath-D+ tumors (62.7%). In multivariate analyses, higher tumor size, no adjuvant chemotherapy and AR/Cath-D co-expression were independent prognostic factors of worse overall survival. Conclusions: AR/Cath-D co-expression independently predicted overall survival. Patients with TNBC in which AR and Cath-D are co-expressed could be eligible for combinatory therapy with androgen antagonists and anti-Cath-D human antibodies.
Collapse
Affiliation(s)
- Hanane Mansouri
- IRCM (Institut de Recherche en Cancérologie de Montpellier), INSERM (Institut National de la Santé et de la Recherche Médicale), Univ Montpellier (University of Montpellier), ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France; (H.M.); (L.B.A.); (A.M.); (W.J.); (V.L.-M.); (P.R.); (S.G.)
| | - Lindsay B. Alcaraz
- IRCM (Institut de Recherche en Cancérologie de Montpellier), INSERM (Institut National de la Santé et de la Recherche Médicale), Univ Montpellier (University of Montpellier), ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France; (H.M.); (L.B.A.); (A.M.); (W.J.); (V.L.-M.); (P.R.); (S.G.)
| | - Caroline Mollevi
- Biometry Department, ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France;
| | - Aude Mallavialle
- IRCM (Institut de Recherche en Cancérologie de Montpellier), INSERM (Institut National de la Santé et de la Recherche Médicale), Univ Montpellier (University of Montpellier), ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France; (H.M.); (L.B.A.); (A.M.); (W.J.); (V.L.-M.); (P.R.); (S.G.)
| | - William Jacot
- IRCM (Institut de Recherche en Cancérologie de Montpellier), INSERM (Institut National de la Santé et de la Recherche Médicale), Univ Montpellier (University of Montpellier), ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France; (H.M.); (L.B.A.); (A.M.); (W.J.); (V.L.-M.); (P.R.); (S.G.)
- Department of Medical Oncology, ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France
- Translational Research Unit, ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France; (F.B.-M.); (J.S.-L.)
| | - Florence Boissière-Michot
- Translational Research Unit, ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France; (F.B.-M.); (J.S.-L.)
| | - Joelle Simony-Lafontaine
- Translational Research Unit, ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France; (F.B.-M.); (J.S.-L.)
| | - Valérie Laurent-Matha
- IRCM (Institut de Recherche en Cancérologie de Montpellier), INSERM (Institut National de la Santé et de la Recherche Médicale), Univ Montpellier (University of Montpellier), ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France; (H.M.); (L.B.A.); (A.M.); (W.J.); (V.L.-M.); (P.R.); (S.G.)
| | - Pascal Roger
- IRCM (Institut de Recherche en Cancérologie de Montpellier), INSERM (Institut National de la Santé et de la Recherche Médicale), Univ Montpellier (University of Montpellier), ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France; (H.M.); (L.B.A.); (A.M.); (W.J.); (V.L.-M.); (P.R.); (S.G.)
- Department of Pathology, CHU (Centre Hospitalier Universitaire) Nîmes, 30029 Nîmes, France
| | - Emmanuelle Liaudet-Coopman
- IRCM (Institut de Recherche en Cancérologie de Montpellier), INSERM (Institut National de la Santé et de la Recherche Médicale), Univ Montpellier (University of Montpellier), ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France; (H.M.); (L.B.A.); (A.M.); (W.J.); (V.L.-M.); (P.R.); (S.G.)
- Correspondence:
| | - Séverine Guiu
- IRCM (Institut de Recherche en Cancérologie de Montpellier), INSERM (Institut National de la Santé et de la Recherche Médicale), Univ Montpellier (University of Montpellier), ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France; (H.M.); (L.B.A.); (A.M.); (W.J.); (V.L.-M.); (P.R.); (S.G.)
- Department of Medical Oncology, ICM (Institut du Cancer de Montpellier), 34298 Montpellier, France
| |
Collapse
|
18
|
Ashraf Y, Mansouri H, Laurent-Matha V, Alcaraz LB, Roger P, Guiu S, Derocq D, Robin G, Michaud HA, Delpech H, Jarlier M, Pugnière M, Robert B, Puel A, Martin L, Landomiel F, Bourquard T, Achour O, Fruitier-Arnaudin I, Pichard A, Deshayes E, Turtoi A, Poupon A, Simony-Lafontaine J, Boissière-Michot F, Pirot N, Bernex F, Jacot W, du Manoir S, Theillet C, Pouget JP, Navarro-Teulon I, Bonnefoy N, Pèlegrin A, Chardès T, Martineau P, Liaudet-Coopman E. Immunotherapy of triple-negative breast cancer with cathepsin D-targeting antibodies. J Immunother Cancer 2019; 7:29. [PMID: 30717773 PMCID: PMC6360707 DOI: 10.1186/s40425-019-0498-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/01/2019] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) treatment is currently restricted to chemotherapy. Hence, tumor-specific molecular targets and/or alternative therapeutic strategies for TNBC are urgently needed. Immunotherapy is emerging as an exciting treatment option for TNBC patients. The aspartic protease cathepsin D (cath-D), a marker of poor prognosis in breast cancer (BC), is overproduced and hypersecreted by human BC cells. This study explores whether cath-D is a tumor cell-associated extracellular biomarker and a potent target for antibody-based therapy in TNBC. METHODS Cath-D prognostic value and localization was evaluated by transcriptomics, proteomics and immunohistochemistry in TNBC. First-in-class anti-cath-D human scFv fragments binding to both human and mouse cath-D were generated using phage display and cloned in the human IgG1 λ format (F1 and E2). Anti-cath-D antibody biodistribution, antitumor efficacy and in vivo underlying mechanisms were investigated in TNBC MDA-MB-231 tumor xenografts in nude mice. Antitumor effect was further assessed in TNBC patient-derived xenografts (PDXs). RESULTS High CTSD mRNA levels correlated with shorter recurrence-free survival in TNBC, and extracellular cath-D was detected in the tumor microenvironment, but not in matched normal breast stroma. Anti-cath-D F1 and E2 antibodies accumulated in TNBC MDA-MB-231 tumor xenografts, inhibited tumor growth and improved mice survival without apparent toxicity. The Fc function of F1, the best antibody candidate, was essential for maximal tumor inhibition in the MDA-MB-231 model. Mechanistically, F1 antitumor response was triggered through natural killer cell activation via IL-15 upregulation, associated with granzyme B and perforin production, and the release of antitumor IFNγ cytokine. The F1 antibody also prevented the tumor recruitment of immunosuppressive tumor-associated macrophages M2 and myeloid-derived suppressor cells, a specific effect associated with a less immunosuppressive tumor microenvironment highlighted by TGFβ decrease. Finally, the antibody F1 inhibited tumor growth of two TNBC PDXs, isolated from patients resistant or not to neo-adjuvant chemotherapy. CONCLUSION Cath-D is a tumor-specific extracellular target in TNBC suitable for antibody-based therapy. Immunomodulatory antibody-based strategy against cath-D is a promising immunotherapy to treat patients with TNBC.
Collapse
Affiliation(s)
- Yahya Ashraf
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Hanane Mansouri
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Valérie Laurent-Matha
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Lindsay B Alcaraz
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Pascal Roger
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
- Department of Pathology, CHU Nîmes, Nîmes, France
| | - Séverine Guiu
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
- Department of Medical Oncology, ICM, Montpellier, France
| | - Danielle Derocq
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Gautier Robin
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Henri-Alexandre Michaud
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Helène Delpech
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | | | - Martine Pugnière
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Bruno Robert
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Anthony Puel
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Lucie Martin
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | | | | | | | | | - Alexandre Pichard
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Emmanuel Deshayes
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Andrei Turtoi
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | | | | | | | - Nelly Pirot
- Réseau d'Histologie Expérimentale de Montpellier, BioCampus, UMS3426 CNRS-US009 INSERM-UM, Montpellier, France
| | - Florence Bernex
- Réseau d'Histologie Expérimentale de Montpellier, BioCampus, UMS3426 CNRS-US009 INSERM-UM, Montpellier, France
| | - William Jacot
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
- Department of Medical Oncology, ICM, Montpellier, France
- Translational Research Unit, ICM, Montpellier, France
| | - Stanislas du Manoir
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Charles Theillet
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Jean-Pierre Pouget
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Isabelle Navarro-Teulon
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Nathalie Bonnefoy
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - André Pèlegrin
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Thierry Chardès
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Pierre Martineau
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France
| | - Emmanuelle Liaudet-Coopman
- IRCM, INSERM, U1194 Univ Montpellier, ICM, 208, rue des Apothicaires, F-34298, Montpellier, Cedex 5, France.
| |
Collapse
|
19
|
Zhang C, Zhang M, Song S. Cathepsin D enhances breast cancer invasion and metastasis through promoting hepsin ubiquitin-proteasome degradation. Cancer Lett 2018; 438:105-115. [PMID: 30227221 DOI: 10.1016/j.canlet.2018.09.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 09/07/2018] [Accepted: 09/11/2018] [Indexed: 11/16/2022]
Abstract
Hepsin is required for the growth and maintenance of normal morphology, as well as for cell motility and development, initiation of blood coagulation and pro-inflammatory immune response. Here we showed that Cathepsin D (CtsD) as a novel protein is involved in the regulation of hepsin. CtsD destabilizes hepsin by promoting its ubiquitylation and subsequent proteasomal degradation in breast cancer cells. Breast cancer tissue microarray also indicated that hepsin expression was negatively correlated with CtsD by immunohistochemistry. Overexpression of CtsD promoted breast cancer cell migration, invasion and metastasis by enhancing the expression of intercellular cell adhesion molecule-1 (ICAM-1) in vitro and in vivo. These effects were inhibited by ectopic hepsin expression. Taken together, our data reveal a critical CtsD-hepsin signaling axis in migration and metastasis, which may contribute to a better understanding of the function and molecular mechanism in breast cancer progression.
Collapse
Affiliation(s)
- Chunyi Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Mingming Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Shushu Song
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| |
Collapse
|
20
|
Overexpression of low density lipoprotein receptor-related protein 1 (LRP1) is associated with worsened prognosis and decreased cancer immunity in clear-cell renal cell carcinoma. Biochem Biophys Res Commun 2018; 503:1537-1543. [PMID: 30033103 DOI: 10.1016/j.bbrc.2018.07.076] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022]
Abstract
AIM Clear-cell renal cell carcinoma (ccRCC) is characterized with underlying genetic disorders and the role of low density lipoprotein receptor-related protein 1 (LRP1) in ccRCC is unknown. METHOD An in silico exploratory analysis using multiple public genetic datasets was used to establish association between LRP1 expression and clinicopathological parameters. Associations of interest were validated using 155 ccRCC samples using immunohistochemistry. RESULTS LRP1 was overexpressed in tumor compared with normal kidney tissue. Increased LRP1 expression in ccRCC was associated with advanced stage, grade and worsened overall survival and progression-free survival. Functional annotation indicated an immune-modulatory role of LRP1 in ccRCC. LRP1 expression was significantly correlated with expressions of PBRM1, SETD2, and KDM5C. Positive correlations between LRP1 and pro-angiogenic factors ERAP1, SCG2, STAB1, and RUNX1 were observed. LRP1 expression was positively correlated with PD-L2 level. Negative correlations between LRP1 and anti-angiogenic factors EMCN and IL18 were observed. LRP1 expression was not associated with microvessel density (MVD) yet was negatively correlated with tumor-infiltrating lymphocytes (TIL). CONCLUSION LRP1 is associated with worsened prognosis in ccRCC and is related to cancer immune modulation. LRP1-targeted therapy can be of therapeutic potential.
Collapse
|
21
|
Oldoni F, van Capelleveen JC, Dalila N, Wolters JC, Heeren J, Sinke RJ, Hui DY, Dallinga-Thie GM, Frikke-Schmidt R, Hovingh KG, van de Sluis B, Tybjærg-Hansen A, Kuivenhoven JA. Naturally Occurring Variants in LRP1 (Low-Density Lipoprotein Receptor-Related Protein 1) Affect HDL (High-Density Lipoprotein) Metabolism Through ABCA1 (ATP-Binding Cassette A1) and SR-B1 (Scavenger Receptor Class B Type 1) in Humans. Arterioscler Thromb Vasc Biol 2018; 38:1440-1453. [PMID: 29853565 PMCID: PMC6023722 DOI: 10.1161/atvbaha.117.310309] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Studies into the role of LRP1 (low-density lipoprotein receptor–related protein 1) in human lipid metabolism are scarce. Although it is known that a common variant in LRP1 (rs116133520) is significantly associated with HDL-C (high-density lipoprotein cholesterol), the mechanism underlying this observation is unclear. In this study, we set out to study the functional effects of 2 rare LRP1 variants identified in subjects with extremely low HDL-C levels. Approach and Results— In 2 subjects with HDL-C below the first percentile for age and sex and moderately elevated triglycerides, we identified 2 rare variants in LRP1: p.Val3244Ile and p.Glu3983Asp. Both variants decrease LRP1 expression and stability. We show in a series of translational experiments that these variants culminate in reduced trafficking of ABCA1 (ATP-binding cassette A1) to the cell membrane. This is accompanied by an increase in cell surface expression of SR-B1 (scavenger receptor class B type 1). Combined these effects may contribute to low HDL-C levels in our study subjects. Supporting these findings, we provide epidemiological evidence that rs116133520 is associated with apo (apolipoprotein) A1 but not with apoB levels. Conclusions— This study provides the first evidence that rare variants in LRP1 are associated with changes in human lipid metabolism. Specifically, this study shows that LRP1 may affect HDL metabolism by virtue of its effect on both ABCA1 and SR-B1.
Collapse
Affiliation(s)
- Federico Oldoni
- From the Department of Pediatrics, Section of Molecular Genetics, University Medical Centre Groningen, University of Groningen, The Netherlands (F.O., J.C.W., B.v.d.S., J.A.K.)
| | | | - Nawar Dalila
- Department of Clinical Biochemistry, Rigshospitalet (N.D., R.F.-S., A.T.-H.)
| | - Justina C Wolters
- From the Department of Pediatrics, Section of Molecular Genetics, University Medical Centre Groningen, University of Groningen, The Netherlands (F.O., J.C.W., B.v.d.S., J.A.K.)
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Germany (J.H.)
| | - Richard J Sinke
- Department of Genetics, University Medical Centre Groningen, The Netherlands (R.J.S.)
| | - David Y Hui
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, OH (D.Y.H.)
| | - Geesje M Dallinga-Thie
- Department of Vascular Medicine (J.C.v.C., G.M.D.-T., K.G.H.).,Department Experimental Vascular Medicine (G.M.D.-T.), Academic Medical Center, Amsterdam, The Netherlands
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet (N.D., R.F.-S., A.T.-H.)
| | - Kees G Hovingh
- Department of Vascular Medicine (J.C.v.C., G.M.D.-T., K.G.H.)
| | - Bart van de Sluis
- From the Department of Pediatrics, Section of Molecular Genetics, University Medical Centre Groningen, University of Groningen, The Netherlands (F.O., J.C.W., B.v.d.S., J.A.K.)
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Rigshospitalet (N.D., R.F.-S., A.T.-H.).,Copenhagen City Heart Study, Frederiksberg Hospital (A.T.-H.), Copenhagen University Hospital and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jan Albert Kuivenhoven
- From the Department of Pediatrics, Section of Molecular Genetics, University Medical Centre Groningen, University of Groningen, The Netherlands (F.O., J.C.W., B.v.d.S., J.A.K.)
| |
Collapse
|
22
|
Markmann S, Krambeck S, Hughes CJ, Mirzaian M, Aerts JMFG, Saftig P, Schweizer M, Vissers JPC, Braulke T, Damme M. Quantitative Proteome Analysis of Mouse Liver Lysosomes Provides Evidence for Mannose 6-phosphate-independent Targeting Mechanisms of Acid Hydrolases in Mucolipidosis II. Mol Cell Proteomics 2017; 16:438-450. [PMID: 28062798 DOI: 10.1074/mcp.m116.063636] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/24/2016] [Indexed: 01/01/2023] Open
Abstract
The efficient receptor-mediated targeting of soluble lysosomal proteins to lysosomes requires the modification with mannose 6-phosphate (M6P) residues. Although the absence of M6P results in misrouting and hypersecretion of lysosomal enzymes in many cells, normal levels of lysosomal enzymes have been reported in liver of patients lacking the M6P-generating phosphotransferase (PT). The identity of lysosomal proteins depending on M6P has not yet been comprehensively analyzed. In this study we purified lysosomes from liver of PT-defective mice and 67 known soluble lysosomal proteins were identified that illustrated quantitative changes using an ion mobility-assisted data-independent label-free LC-MS approach. After validation of various differentially expressed lysosomal components by Western blotting and enzyme activity assays, the data revealed a small number of lysosomal proteins depending on M6P, including neuraminidase 1, cathepsin F, Npc2, and cathepsin L, whereas the majority reach lysosomes by alternative pathways. These data were compared with findings on cultured hepatocytes and liver sinusoid endothelial cells isolated from the liver of wild-type and PT-defective mice. Our findings show that the relative expression, targeting efficiency and lysosomal localization of lysosomal proteins tested in cultured hepatic cells resemble their proportion in isolated liver lysosomes. Hypersecretion of newly synthesized nonphosphorylated lysosomal proteins suggest that secretion-recapture mechanisms contribute to maintain major lysosomal functions in liver.
Collapse
Affiliation(s)
- Sandra Markmann
- From the ‡Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,§Waters Corporation, Wilmslow, SK9 4AX, United Kingdom
| | - Svenja Krambeck
- From the ‡Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,§Waters Corporation, Wilmslow, SK9 4AX, United Kingdom
| | | | - Mina Mirzaian
- ¶Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, 2333 CC, The Netherlands
| | - Johannes M F G Aerts
- ¶Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, 2333 CC, The Netherlands
| | - Paul Saftig
- ‖Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24098 Kiel, Germany
| | - Michaela Schweizer
- **Morphology Unit, Center for Molecular Neurobiology ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Thomas Braulke
- From the ‡Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany;
| | - Markus Damme
- ‖Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24098 Kiel, Germany;
| |
Collapse
|
23
|
Subcellular Trafficking of Mammalian Lysosomal Proteins: An Extended View. Int J Mol Sci 2016; 18:ijms18010047. [PMID: 28036022 PMCID: PMC5297682 DOI: 10.3390/ijms18010047] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/15/2016] [Accepted: 12/18/2016] [Indexed: 01/02/2023] Open
Abstract
Lysosomes clear macromolecules, maintain nutrient and cholesterol homeostasis, participate in tissue repair, and in many other cellular functions. To assume these tasks, lysosomes rely on their large arsenal of acid hydrolases, transmembrane proteins and membrane-associated proteins. It is therefore imperative that, post-synthesis, these proteins are specifically recognized as lysosomal components and are correctly sorted to this organelle through the endosomes. Lysosomal transmembrane proteins contain consensus motifs in their cytosolic regions (tyrosine- or dileucine-based) that serve as sorting signals to the endosomes, whereas most lysosomal acid hydrolases acquire mannose 6-phosphate (Man-6-P) moieties that mediate binding to two membrane receptors with endosomal sorting motifs in their cytosolic tails. These tyrosine- and dileucine-based motifs are tickets for boarding in clathrin-coated carriers that transport their cargo from the trans-Golgi network and plasma membrane to the endosomes. However, increasing evidence points to additional mechanisms participating in the biogenesis of lysosomes. In some cell types, for example, there are alternatives to the Man-6-P receptors for the transport of some acid hydrolases. In addition, several “non-consensus” sorting motifs have been identified, and atypical transport routes to endolysosomes have been brought to light. These “unconventional” or “less known” transport mechanisms are the focus of this review.
Collapse
|
24
|
O'Donoghue AJ, Ivry SL, Chaudhury C, Hostetter DR, Hanahan D, Craik CS. Procathepsin E is highly abundant but minimally active in pancreatic ductal adenocarcinoma tumors. Biol Chem 2016; 397:871-81. [PMID: 27149201 PMCID: PMC5712230 DOI: 10.1515/hsz-2016-0138] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/20/2016] [Indexed: 12/31/2022]
Abstract
The cathepsin family of lysosomal proteases is increasingly being recognized for their altered expression in cancer and role in facilitating tumor progression. The aspartyl protease cathepsin E is overexpressed in several cancers and has been investigated as a biomarker for pancreatic ductal adenocarcinoma (PDAC). Here we show that cathepsin E expression in mouse PDAC tumors is increased by more than 400-fold when compared to healthy pancreatic tissue. Cathepsin E accumulates over the course of disease progression and accounts for more than 3% of the tumor protein in mice with end-stage disease. Through immunoblot analysis we determined that only procathepsin E exists in mouse PDAC tumors and cell lines derived from these tumors. By decreasing the pH, this procathepsion E is converted to the mature form, resulting in an increase in proteolytic activity. Although active site inhibitors can bind procathepsin E, treatment of PDAC mice with the aspartyl protease inhibitor ritonavir did not decrease tumor burden. Lastly, we used multiplex substrate profiling by mass spectrometry to identify two synthetic peptides that are hydrolyzed by procathepsin E near neutral pH. This work represents a comprehensive analysis of procathepsin E in PDAC and could facilitate the development of improved biomarkers for disease detection.
Collapse
|
25
|
Double-targeted polymersomes and liposomes for multiple barrier crossing. Int J Pharm 2016; 511:946-56. [DOI: 10.1016/j.ijpharm.2016.08.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 07/31/2016] [Accepted: 08/02/2016] [Indexed: 01/09/2023]
|
26
|
Bach AS, Derocq D, Laurent-Matha V, Montcourrier P, Sebti S, Orsetti B, Theillet C, Gongora C, Pattingre S, Ibing E, Roger P, Linares LK, Reinheckel T, Meurice G, Kaiser FJ, Gespach C, Liaudet-Coopman E. Nuclear cathepsin D enhances TRPS1 transcriptional repressor function to regulate cell cycle progression and transformation in human breast cancer cells. Oncotarget 2016; 6:28084-103. [PMID: 26183398 PMCID: PMC4695046 DOI: 10.18632/oncotarget.4394] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/15/2015] [Indexed: 11/25/2022] Open
Abstract
The lysosomal protease cathepsin D (Cath-D) is overproduced in breast cancer cells (BCC) and supports tumor growth and metastasis formation. Here, we describe the mechanism whereby Cath-D is accumulated in the nucleus of ERα-positive (ER+) BCC. We identified TRPS1 (tricho-rhino-phalangeal-syndrome 1), a repressor of GATA-mediated transcription, and BAT3 (Scythe/BAG6), a nucleo-cytoplasmic shuttling chaperone protein, as new Cath-D-interacting nuclear proteins. Cath-D binds to BAT3 in ER+ BCC and they partially co-localize at the surface of lysosomes and in the nucleus. BAT3 silencing inhibits Cath-D accumulation in the nucleus, indicating that Cath-D nuclear targeting is controlled by BAT3. Fully mature Cath-D also binds to full-length TRPS1 and they co-localize in the nucleus of ER+ BCC where they are associated with chromatin. Using the LexA-VP16 fusion co-activator reporter assay, we then show that Cath-D acts as a transcriptional repressor, independently of its catalytic activity. Moreover, microarray analysis of BCC in which Cath-D and/or TRPS1 expression were silenced indicated that Cath-D enhances TRPS1-mediated repression of several TRPS1-regulated genes implicated in carcinogenesis, including PTHrP, a canonical TRPS1 gene target. In addition, co-silencing of TRPS1 and Cath-D in BCC affects the transcription of cell cycle, proliferation and transformation genes, and impairs cell cycle progression and soft agar colony formation. These findings indicate that Cath-D acts as a nuclear transcriptional cofactor of TRPS1 to regulate ER+ BCC proliferation and transformation in a non-proteolytic manner.
Collapse
Affiliation(s)
- Anne-Sophie Bach
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Danielle Derocq
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Valérie Laurent-Matha
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Philippe Montcourrier
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Salwa Sebti
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Béatrice Orsetti
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Charles Theillet
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Céline Gongora
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Sophie Pattingre
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Eva Ibing
- Universität zu Lübeck, Lübeck, Germany
| | - Pascal Roger
- Department of Pathology, CHU Nimes, Nimes, France
| | - Laetitia K Linares
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany
| | - Guillaume Meurice
- Functional Genomic Plateform, Institut Gustave Roussy, Villejuif, France
| | | | - Christian Gespach
- INSERM U938, Molecular and Clinical Oncology, Paris 6 University Pierre et Marie Curie, Hôpital Saint-Antoine, Paris, France
| | - Emmanuelle Liaudet-Coopman
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France.,INSERM U1194, Montpellier, France.,Université de Montpellier, Montpellier, France.,Institut Régional du Cancer de Montpellier, Montpellier, France
| |
Collapse
|
27
|
Boonen M, Staudt C, Gilis F, Oorschot V, Klumperman J, Jadot M. Cathepsin D and its newly identified transport receptor SEZ6L2 can modulate neurite outgrowth. J Cell Sci 2015; 129:557-68. [PMID: 26698217 DOI: 10.1242/jcs.179374] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 12/17/2015] [Indexed: 12/14/2022] Open
Abstract
How, in the absence of a functional mannose 6-phosphate (Man-6-P)-signal-dependent transport pathway, some acid hydrolases remain sorted to endolysosomes in the brain is poorly understood. We demonstrate that cathepsin D binds to mouse SEZ6L2, a type 1 transmembrane protein predominantly expressed in the brain. Studies of the subcellular trafficking of SEZ6L2, and its silencing in a mouse neuroblastoma cell line reveal that SEZ6L2 is involved in the trafficking of cathepsin D to endosomes. Moreover, SEZ6L2 can partially correct the cathepsin D hypersecretion resulting from the knockdown of UDP-GlcNAc:lysosomal enzyme GlcNAc-1-phosphotransferase in HeLa cells (i.e. in cells that are unable to synthesize Man-6-P signals). Interestingly, cleavage of SEZ6L2 by cathepsin D generates an N-terminal soluble fragment that induces neurite outgrowth, whereas its membrane counterpart prevents this. Taken together, our findings highlight that SEZ6L2 can serve as receptor to mediate the sorting of cathepsin D to endosomes, and suggest that proteolytic cleavage of SEZ6L2 by cathepsin D modulates neuronal differentiation.
Collapse
Affiliation(s)
- Marielle Boonen
- URPhyM-Laboratoire de Chimie Physiologique, University of Namur, 61 rue de Bruxelles, Namur 5000, Belgium
| | - Catherine Staudt
- URPhyM-Laboratoire de Chimie Physiologique, University of Namur, 61 rue de Bruxelles, Namur 5000, Belgium
| | - Florentine Gilis
- URPhyM-Laboratoire de Chimie Physiologique, University of Namur, 61 rue de Bruxelles, Namur 5000, Belgium
| | - Viola Oorschot
- Department of Cell Biology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, The Netherlands
| | - Judith Klumperman
- Department of Cell Biology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, The Netherlands
| | - Michel Jadot
- URPhyM-Laboratoire de Chimie Physiologique, University of Namur, 61 rue de Bruxelles, Namur 5000, Belgium
| |
Collapse
|
28
|
Van Gool B, Dedieu S, Emonard H, Roebroek AJM. The Matricellular Receptor LRP1 Forms an Interface for Signaling and Endocytosis in Modulation of the Extracellular Tumor Environment. Front Pharmacol 2015; 6:271. [PMID: 26617523 PMCID: PMC4639618 DOI: 10.3389/fphar.2015.00271] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/29/2015] [Indexed: 12/19/2022] Open
Abstract
The membrane protein low-density lipoprotein receptor related-protein 1 (LRP1) has been attributed a role in cancer. However, its presumably often indirect involvement is far from understood. LRP1 has both endocytic and signaling activities. As a matricellular receptor it is involved in regulation, mostly by clearing, of various extracellular matrix degrading enzymes including matrix metalloproteinases, serine proteases, protease inhibitor complexes, and the endoglycosidase heparanase. Furthermore, by binding extracellular ligands including growth factors and subsequent intracellular interaction with scaffolding and adaptor proteins it is involved in regulation of various signaling cascades. LRP1 expression levels are often downregulated in cancer and some studies consider low LRP1 levels a poor prognostic factor. On the contrary, upregulation in brain cancers has been noted and clinical trials explore the use of LRP1 as cargo receptor to deliver cytotoxic agents. This mini-review focuses on LRP1's role in tumor growth and metastasis especially by modulation of the extracellular tumor environment. In relation to this role its diagnostic, prognostic and therapeutic potential will be discussed.
Collapse
Affiliation(s)
- Bart Van Gool
- Laboratory for Experimental Mouse Genetics, Department of Human Genetics , KU Leuven, Leuven, Belgium
| | - Stéphane Dedieu
- 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
| | - Hervé Emonard
- 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
| | - Anton J M Roebroek
- Laboratory for Experimental Mouse Genetics, Department of Human Genetics , KU Leuven, Leuven, Belgium
| |
Collapse
|
29
|
Thevenard J, Verzeaux L, Devy J, Etique N, Jeanne A, Schneider C, Hachet C, Ferracci G, David M, Martiny L, Charpentier E, Khrestchatisky M, Rivera S, Dedieu S, Emonard H. Low-density lipoprotein receptor-related protein-1 mediates endocytic clearance of tissue inhibitor of metalloproteinases-1 and promotes its cytokine-like activities. PLoS One 2014; 9:e103839. [PMID: 25075518 PMCID: PMC4116228 DOI: 10.1371/journal.pone.0103839] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 06/11/2014] [Indexed: 11/19/2022] Open
Abstract
Tissue inhibitor of metalloproteinases-1 (TIMP-1) regulates the extracellular matrix turnover by inhibiting the proteolytic activity of matrix metalloproteinases (MMPs). TIMP-1 also displays MMP-independent activities that influence the behavior of various cell types including neuronal plasticity, but the underlying molecular mechanisms remain mostly unknown. The trans-membrane receptor low-density lipoprotein receptor-related protein-1 (LRP-1) consists of a large extracellular chain with distinct ligand-binding domains that interact with numerous ligands including TIMP-2 and TIMP-3 and a short transmembrane chain with intracellular motifs that allow endocytosis and confer signaling properties to LRP-1. We addressed TIMP-1 interaction with recombinant ligand-binding domains of LRP-1 expressed by CHO cells for endocytosis study, or linked onto sensor chips for surface plasmon resonance analysis. Primary cortical neurons bound and internalized endogenous TIMP-1 through a mechanism mediated by LRP-1. This resulted in inhibition of neurite outgrowth and increased growth cone volume. Using a mutated inactive TIMP-1 variant we showed that TIMP-1 effect on neurone morphology was independent of its MMP inhibitory activity. We conclude that TIMP-1 is a new ligand of LRP-1 and we highlight a new example of its MMP-independent, cytokine-like functions.
Collapse
Affiliation(s)
- Jessica Thevenard
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims-Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Sciences Exactes et Naturelles, Reims, France
| | - Laurie Verzeaux
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims-Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Sciences Exactes et Naturelles, Reims, France
| | - Jerôme Devy
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims-Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Sciences Exactes et Naturelles, Reims, France
| | - Nicolas Etique
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims-Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Sciences Exactes et Naturelles, Reims, France
| | - Albin Jeanne
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims-Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Sciences Exactes et Naturelles, Reims, France
| | - Christophe Schneider
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims-Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Sciences Exactes et Naturelles, Reims, France
| | - Cathy Hachet
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims-Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Sciences Exactes et Naturelles, Reims, France
| | - Géraldine Ferracci
- Aix-Marseille Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M), UMR 7286, Plate-Forme de Recherche en Neurosciences (PFRN), Marseille, France
| | - Marion David
- VECT-HORUS SAS, Faculté de Médecine Secteur Nord, Marseille, France
| | - Laurent Martiny
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims-Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Sciences Exactes et Naturelles, Reims, France
| | - Emmanuelle Charpentier
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims-Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Sciences Exactes et Naturelles, Reims, France
| | - Michel Khrestchatisky
- Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, Aix-Marseille Université, Marseille, France
- NICN, CNRS UMR 7259, Marseille, France
| | - Santiago Rivera
- Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 7259, Aix-Marseille Université, Marseille, France
- NICN, CNRS UMR 7259, Marseille, France
| | - Stéphane Dedieu
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims-Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Sciences Exactes et Naturelles, Reims, France
| | - Hervé Emonard
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7369 Matrice Extracellulaire et Dynamique Cellulaire, Université de Reims-Champagne-Ardenne, Unité de Formation et de Recherche (UFR) Sciences Exactes et Naturelles, Reims, France
- * E-mail:
| |
Collapse
|
30
|
Khalkhali-Ellis Z, Goossens W, Margaryan NV, Hendrix MJC. Cleavage of Histone 3 by Cathepsin D in the involuting mammary gland. PLoS One 2014; 9:e103230. [PMID: 25054204 PMCID: PMC4108390 DOI: 10.1371/journal.pone.0103230] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 06/30/2014] [Indexed: 01/08/2023] Open
Abstract
The post-lactational regression of mammary gland is a complex multi-step process designed to conserve the biological function of the gland for next pregnancy. This developmental stage is a biological intrigue with great relevance to breast cancer research, and thus has been the subject of intensive scrutiny. Multipronged studies (microarray, proteomics profiling, animal knock-out models) have provided a repertoire of genes critical to involution. However, the caveat of these approaches remains in their failure to reveal post-translational modification(s), an emerging and critical aspect of gene regulation in developmental processes and mammary gland remodeling. The massive surge in the lysosomal enzymes concurrent with the onset of involution has been known for decades, and considered essential for “clearance” purposes. However, functional significance of these enzymes in diverse biological processes distinct from their proteolytic activity is just emerging. Studies from our laboratory had indicated specific post-translational modifications of the aspartyl endopeptidase Cathepsin D (CatD) at distinct stages mammary gland development. This study addresses the biological significance of these modifications in the involution process, and reveals that post-translational modifications drive CatD into the nucleus to cleave Histone 3. The cleavage of Histone 3 has been associated with cellular differentiation and could be critical instigator of involution process. From functional perspective, deregulated expression and increased secretion of CatD are associated with aggressive and metastatic phenotype of breast cancer. Thus unraveling CatD’s physiological functions in mammary gland development will bridge the present gap in understanding its pro-tumorigenic/metastatic functions, and assist in the generation of tailored therapeutic approaches.
Collapse
Affiliation(s)
- Zhila Khalkhali-Ellis
- Cancer Biology and Epigenomics, Stanley Manne Children’s Research Institute, Chicago, Illinois, United States of America
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- * E-mail:
| | - William Goossens
- Cancer Biology and Epigenomics, Stanley Manne Children’s Research Institute, Chicago, Illinois, United States of America
| | - Naira V. Margaryan
- Cancer Biology and Epigenomics, Stanley Manne Children’s Research Institute, Chicago, Illinois, United States of America
| | - Mary J. C. Hendrix
- Cancer Biology and Epigenomics, Stanley Manne Children’s Research Institute, Chicago, Illinois, United States of America
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| |
Collapse
|
31
|
Khalkhali-Ellis Z, Hendrix MJC. Two Faces of Cathepsin D: Physiological Guardian Angel and Pathological Demon. ACTA ACUST UNITED AC 2014; 6. [PMID: 25663755 PMCID: PMC4318633 DOI: 10.4172/0974-8369.1000206] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Since its discovery as a lysosomal hydrolase, Cathepsin D (CatD) has been the subject of intensive scrutiny by numerous scientists. Those accumulated efforts have defined its biosynthetic pathway, structure, and companion proteins in the context of its perceived “house keeping” function. However, in the past two decades CatD has emerged as a multifunctional enzyme, involved in myriad biological processes beyond its original “housekeeping” role. CatD is responsible for selective and limited cleavage (quite distinct from non-specific protein degradation) of particular substrates vital to proper cellular function. These proteolytic events are critical in the control of biological processes, including cell cycle progression, differentiation and migration, morphogenesis and tissue remodeling, immunological processes, ovulation, fertilization, neuronal outgrowth, angiogenesis, and apoptosis. Consistent with the biological relevance of CatD, its deficiency, altered regulation or post-translational modification underlie important pathological conditions such as cancer, atherosclerosis, neurological and skin disorders. Specifically, deregulated synthesis, post-translational modifications and hyper-secretion of CatD, along with its mitogenic effects, are established hallmarks of cancer. More importantly, but less studied, is its significance in regulating the sensitivity to anticancer drugs. This review outlines CatD’s post-translational modifications, cellular trafficking, secretion and protein binding partners in normal mammary gland, and restates the “site-specific” function of CatD which is most probably dictated by its post-translational modifications and binding partners. Noteworthy, CatD’s association with one of its binding partners in the context of drug sensitivity is highlighted, with the optimism that it could contribute to the development of more effective chemotherapeutic agent(s) tailored for individual patients.
Collapse
Affiliation(s)
- Zhila Khalkhali-Ellis
- Stanley Manne Children's Research Institute, Northwestern University Feinberg School of Medicine, 2300 Children's Plaza, Box 222, Chicago, Illinois, 60614-3394, USA
| | - Mary J C Hendrix
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 2300 Children's Plaza, Box 222, Chicago, Illinois, 60614-3394, USA
| |
Collapse
|
32
|
Human herpesvirus 8 interleukin-6 contributes to primary effusion lymphoma cell viability via suppression of proapoptotic cathepsin D, a cointeraction partner of vitamin K epoxide reductase complex subunit 1 variant 2. J Virol 2013; 88:1025-38. [PMID: 24198402 DOI: 10.1128/jvi.02830-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human herpesvirus 8 (HHV-8) interleukin-6 (vIL-6) promotes cell proliferation and survival and is proangiogenic, implicating it as a contributor to virus-associated Kaposi's sarcoma, primary effusion lymphoma (PEL), and multicentric Castleman's disease. Although predominantly lytically expressed, vIL-6 is also produced at low, functional levels during latency in PEL cells. Unlike other IL-6 cytokines, vIL-6 is secreted very inefficiently and localizes in the endoplasmic reticulum (ER). ER-localized vIL-6 supports PEL cell proliferation and survival, mediated in part through its interaction with the largely uncharacterized ER-resident protein vitamin K epoxide reductase complex subunit 1 variant 2 (VKORC1v2). Here, we report that the ER-transiting and functionally mitogenic secreted proenzyme (pCatD) form of cathepsin D (mature CatD), a proapoptotic lysosomal aspartate protease, is an interaction partner of VKORC1v2 and that vIL-6 promotes this interaction. Depletion of vIL-6 in PEL cells increased levels of the catalytically active, proteolytically cleaved form of CatD, corresponding with decreased PEL cell viability. Ectopic expression of CatD in PEL cells induced apoptosis, suggesting that CatD suppression by vIL-6 is biologically significant. In the context of high-density culture or reactivation of HHV-8 lytic replication in PEL cells, CatD depletion substantially reduced stress-induced apoptosis and increased virus production. In contrast, CatD overexpression, vIL-6 depletion, and peptide-mediated disruption of vIL-6-VKORC1v2 interaction inhibited replication and cell survival. Combined, our data identify pCatD as an interaction partner of VKORC1v2, demonstrate a role of vIL-6 in CatD suppression via VKORC1v2 in PEL cells, and identify a biologically significant mechanism of vIL-6 prosurvival and proreplication activities via VKORC1v2.
Collapse
|
33
|
Maynadier M, Farnoud R, Lamy PJ, Laurent-Matha V, Garcia M, Rochefort H. Cathepsin D stimulates the activities of secreted plasminogen activators in the breast cancer acidic environment. Int J Oncol 2013; 43:1683-90. [PMID: 24026424 DOI: 10.3892/ijo.2013.2095] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 06/26/2013] [Indexed: 11/05/2022] Open
Abstract
Two proteases cathepsin D (cath D) and urokinase plasminogen activator (uPA) are tissue markers associated with an increased risk of metastasis in breast cancer. We investigated whether cath D, the major aspartyl protease overexpressed by breast cancer cells can trigger a proteolytic cascade via activation of plasminogens at the extracellular pH measured in hypoxic tumors. The effects of the aspartyl protease inhibitor pepstatin on the plasminogen activator (PA) system were analysed by conditioning media of human MDA-MB231 breast cancer cells at pH 6.6 and pH 7.4. Zymography analysis of culture media showed that pepstatin inhibited the secreted activity of tissue-type plasminogen activator (tPA) but not that of uPA. tPA was identified on the basis of the molecular weight, the immunoreactivity with relevant antibodies and the resistance to amiloride, a specific uPA inhibitor. The secreted tPA activity measured by a chromogenic assay in the presence of amiloride was also inhibited by pepstatin at pH 6.6. Surprisingly, pepstatin did not affect secreted tPA protein concentration but markedly increased the amount of the secreted plasminogen activator inhibitor-1 (PAI-1). We conclude that cath D overexpressed by these cells, stimulates at pH 6.6, but not at neutral pH, the extracellular PA proteolytic activity indirectly via PAI-1 proteolysis. This suggests that cath D at acidic pH close to the hypoxic regions of solid tumors, contributes to trigger a proteolytic cascade facilitating cancer cell invasion and metastasis.
Collapse
Affiliation(s)
- Marie Maynadier
- IBMM UMR 5247, University of Montpellier 1, University of Montpellier 2, ENSCM, Montpellier, France
| | | | | | | | | | | |
Collapse
|
34
|
Achour O, Bridiau N, Kacem M, Delatouche R, Bordenave-Juchereau S, Sannier F, Thiéry V, Piot JM, Maugard T, Arnaudin I. Cathepsin D activity and selectivity in the acidic conditions of a tumor microenvironment: Utilization in the development of a novel Cathepsin D substrate for simultaneous cancer diagnosis and therapy. Biochimie 2013; 95:2010-7. [PMID: 23871913 DOI: 10.1016/j.biochi.2013.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 07/08/2013] [Indexed: 12/11/2022]
Abstract
Pro-Cathepsin D (pCD) is an aspartyl endopeptidase which is over expressed in many cancers. This over expression generally led to its secretion into the extracellular culture medium of cancer cells. Moreover, pCD can auto activate and cleave its substrates at an acidic pH compatible with that found in tumor microenvironments (TME). Thus, exploiting these two pathological characteristics of TME offers the opportunity to develop new protease-activated vector on the basis of their specific substrate structures. The aim of this study was to validate new pCD substrates in the extracellular pH conditions of TME. As a first step, we investigated the effect of pH on the catalytic activity and selectivity of mature Cathepsin D (CD). It was found that the increase in the pH of the media led to a decrease in the reaction rate. However, the specificity of mature CD was not affected by a variation in pH. In the second step, the effect of the substrate structure was studied. We demonstrated that the substrate structure had a significant effect on the catalytic activity of CD. In fact, some modifications in peptide structure induced a change in the catalytic behavior that involved a substrate activation phenomenon. We suggest that this activation may be related to the amphiphilic nature of the modified peptide that may induce an interfacial activation mechanism. Finally, pCD, which is the major form found in the extracellular culture medium of cancer cells, was used. We demonstrated that the proform of CD cleave the modified peptide 5 at pH 6.5 with the same cleavage selectivity obtained with the mature form of the protease. These data provide a better understanding of CD behavior in tumor microenvironment conditions and this knowledge can be used to develop more specific tools for diagnosis and drug delivery.
Collapse
Affiliation(s)
- Oussama Achour
- Université de La Rochelle, UMR CNRS 7266, LIENSs, Equipe Approches Moléculaires, Environnement-Santé, Département de Biotechnologies, Avenue Michel Crépeau, 17042 La Rochelle, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
LRP-1: a checkpoint for the extracellular matrix proteolysis. BIOMED RESEARCH INTERNATIONAL 2013; 2013:152163. [PMID: 23936774 PMCID: PMC3723059 DOI: 10.1155/2013/152163] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 06/20/2013] [Indexed: 11/29/2022]
Abstract
Low-density lipoprotein receptor-related protein-(LRP-1) is a large endocytic receptor that binds more than 35 ligands and exhibits signaling properties. Proteinases capable of degrading extracellular matrix (ECM), called matrix proteinases in this paper, are mainly serine proteinases: the activators of plasminogen into plasmin, tissue-type (tPA) and urokinase-type (uPA) plasminogen activators, and the members of the matrix metalloproteinase (MMP) family. LRP-1 is responsible for clearing matrix proteinases, complexed or not with inhibitors. This paper attempts to summarize some aspects on the cellular and molecular bases of endocytic and signaling functions of LRP-1 that modulate extra- and pericellular levels of matrix proteinases.
Collapse
|
36
|
The mannose 6-phosphate-binding sites of M6P/IGF2R determine its capacity to suppress matrix invasion by squamous cell carcinoma cells. Biochem J 2013; 451:91-9. [PMID: 23347038 PMCID: PMC3632087 DOI: 10.1042/bj20121422] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The M6P (mannose 6-phosphate)/IGF2R (insulin-like growth factor II receptor) interacts with a
variety of factors that impinge on tumour invasion and metastasis. It has been shown that expression
of wild-type M6P/IGF2R reduces the tumorigenic and invasive properties of receptor-deficient SCC-VII
squamous cell carcinoma cells. We have now used mutant forms of M6P/IGF2R to assess the relevance of
the different ligand-binding sites of the receptor for its biological activities in this cellular
system. The results of the present study demonstrate that M6P/IGF2R does not require a functional
binding site for insulin-like growth factor II for inhibition of anchorage-independent growth and
matrix invasion by SCC-VII cells. In contrast, the simultaneous mutation of both M6P-binding sites
is sufficient to impair all cellular functions of the receptor tested. These findings highlight that
the interaction between M6P/IGF2R and M6P-modified ligands is not only important for intracellular
accumulation of lysosomal enzymes and formation of dense lysosomes, but is also crucial for the
ability of the receptor to suppress SCC-VII growth and invasion. The present study also shows that
some of the biological activities of M6P/IGF2R in SCC-VII cells strongly depend on a functional
M6P-binding site within domain 3, thus providing further evidence for the non-redundant cellular
functions of the individual carbohydrate-binding domains of the receptor.
Collapse
|
37
|
McConnell RM, Inapudi K, Kadasala N, Yarlagadda K, Velusamy P, McConnell MS, Green A, Trana C, Sayyar K, McConnell JS. New cathepsin D inhibitor library utilizing hydroxyethyl isosteres with cyclic tertiary amines. Med Chem 2013; 8:1146-54. [PMID: 22830497 DOI: 10.2174/1573406411208061146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 05/10/2012] [Accepted: 05/21/2012] [Indexed: 12/23/2022]
Abstract
The design and synthesis of hydroxyethylamine isosteres as inhibitors of cathepsin D based on SAR data have been accomplished. A library of 96 of these hydroxyethylamine isosteres are described and many have proven to be very potent inhibitors of human cathepsin D activity as measured using a fluorometric assay technique, via peptide substrate Ac-Glu-Glu(Edans)-Lys-Pro-Ile-Cys-Phe-Phe-Arg-Leu-Gly-Lys(Methyl Red)-Glu-NH(2). Compounds showing strongest inhibition of cathepsin D activity were those that contain a hydroxyethyl-N'-2- or N'-(4-chlorophenyl)piperazine moiety (IC(50) values range from 0.55 to 8.5 nM), with N'-(2-pyrimidyl)piperizine (IC(50) values range from 0.5 to 21.6 nM), with N-N'- L-piperazinocolinamide (IC(50) values range from 0.001 - 0.25 nM), or N-N'-L-piperazinocolin-N-methylamide (IC(50) values range from 0.015 - 7.3 nM).
Collapse
Affiliation(s)
- Rose M McConnell
- Department of Chemistry, Western Illinois University, Macomb, IL 61455, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
The molecular mechanisms of antimetastatic ruthenium compounds explored through DIGE proteomics. J Inorg Biochem 2013; 118:94-9. [DOI: 10.1016/j.jinorgbio.2012.10.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 10/02/2012] [Accepted: 10/04/2012] [Indexed: 01/04/2023]
|
39
|
Vetvicka V, Fusek M, Vashishta A. Procathepsin d involvement in chemoresistance of cancer cells. NORTH AMERICAN JOURNAL OF MEDICAL SCIENCES 2012; 4:174-9. [PMID: 22536560 PMCID: PMC3334257 DOI: 10.4103/1947-2714.94943] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Background: The role of pCD in cancer has been studied for a long time. We have focused on the hypothesis that increased expression and/or secretion of pCD in cancer cells causes increased chemoresistance to apoptosis inducing molecules. Aim: The aim was to evaluate the effects of pCD expression/release on chemoresistance. Materials and Methods: We tested the LC50 values for various transfectants of breast cancer cell line MDA-MB-231 as well as effects of exogenous additions of pCD, its mutants, pepstatine, antibodies, and Brefeldin on the resistance. Results: We found that pCD levels can be correlated with chemoresistance, the pro-resistant activity seems to be localized outside the cells, proteolytic activity is not involved, and PI3-Akt signaling has an important role in antiapoptotic effects of pCD. Conclusion: We can conclude that overexpression of pCD has strong influence on increased resistance of tumor cells. This could, in fact, be an important contribution in the possible use of pCD level determination for prognostic and/or therapeutic purposes.
Collapse
Affiliation(s)
- Vaclav Vetvicka
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | | | | |
Collapse
|
40
|
Laurent‐Matha V, Huesgen PF, Masson O, Derocq D, Prébois C, Gary‐Bobo M, Lecaille F, Rebière B, Meurice G, Oréar C, Hollingsworth RE, Abrahamson M, Lalmanach G, Overall CM, Liaudet‐Coopman E. Proteolysis of cystatin C by cathepsin D in the breast cancer microenvironment. FASEB J 2012; 26:5172-81. [DOI: 10.1096/fj.12-205229] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Valérie Laurent‐Matha
- Institut de Recherche en Cancérologie de Montpellier (IRCM)Institut National de la Santé et de la Recherche Médicale (INSERM) U896, Université Montpellier1MontpellierFrance
- Centre Régional de Lutte Contre le Cancer (CRLC) Val d'Aurelle Paul LamarqueMontpellierFrance
| | - Pitter F. Huesgen
- Centre for Blood ResearchUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Olivier Masson
- Institut de Recherche en Cancérologie de Montpellier (IRCM)Institut National de la Santé et de la Recherche Médicale (INSERM) U896, Université Montpellier1MontpellierFrance
- Centre Régional de Lutte Contre le Cancer (CRLC) Val d'Aurelle Paul LamarqueMontpellierFrance
| | - Danielle Derocq
- Institut de Recherche en Cancérologie de Montpellier (IRCM)Institut National de la Santé et de la Recherche Médicale (INSERM) U896, Université Montpellier1MontpellierFrance
- Centre Régional de Lutte Contre le Cancer (CRLC) Val d'Aurelle Paul LamarqueMontpellierFrance
| | - Christine Prébois
- Institut de Recherche en Cancérologie de Montpellier (IRCM)Institut National de la Santé et de la Recherche Médicale (INSERM) U896, Université Montpellier1MontpellierFrance
- Centre Régional de Lutte Contre le Cancer (CRLC) Val d'Aurelle Paul LamarqueMontpellierFrance
| | - Magali Gary‐Bobo
- Institut de Recherche en Cancérologie de Montpellier (IRCM)Institut National de la Santé et de la Recherche Médicale (INSERM) U896, Université Montpellier1MontpellierFrance
- Centre Régional de Lutte Contre le Cancer (CRLC) Val d'Aurelle Paul LamarqueMontpellierFrance
| | - Fabien Lecaille
- INSERM U1100, Pathologies Respiratoires: Protéolyse et Aérosolthérapie, Faculté de MédecineUniversité François RabelaisToursFrance
| | - Bertrand Rebière
- Centre de Recherche de Biochimie Macromoléculaire (CRBM)Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 5237MontpellierFrance
| | | | - Cédric Oréar
- Functional Genomic PlatformInstitut Gustave RoussyVillejuifFrance
| | | | - Magnus Abrahamson
- Department of Laboratory Medicine, Division of Clinical ChemistryLund UniversityUniversity HospitalLundSweden
| | - Gilles Lalmanach
- INSERM U1100, Pathologies Respiratoires: Protéolyse et Aérosolthérapie, Faculté de MédecineUniversité François RabelaisToursFrance
| | | | - Emmanuelle Liaudet‐Coopman
- Institut de Recherche en Cancérologie de Montpellier (IRCM)Institut National de la Santé et de la Recherche Médicale (INSERM) U896, Université Montpellier1MontpellierFrance
- Centre Régional de Lutte Contre le Cancer (CRLC) Val d'Aurelle Paul LamarqueMontpellierFrance
| |
Collapse
|
41
|
Abstract
The low-density lipoprotein receptor-related protein 1 (LRP-1) is a large endocytic receptor mediating the clearance of various molecules from the extracellular matrix. In the field of cancer, LRP-1-mediated endocytosis was first associated with antitumor properties. However, recent results suggested that LRP-1 may coordinate the adhesion-deadhesion balance in malignant cells to support tumor progression. Here, we observed that LRP-1 silencing or RAP (receptor-associated protein) treatment led to accumulation of CD44 at the tumor cell surface. Moreover, we evidenced a tight interaction between CD44 and LRP-1, not exclusively localized in lipid rafts. Overexpression of LRP-1-derived minireceptors indicated that the fourth ligand-binding cluster of LRP-1 is required to bind CD44. Labeling of CD44 with EEA1 and LAMP-1 showed that internalized CD44 is routed through early endosomes toward lysosomes in a LRP-1-dependent pathway. LRP-1-mediated internalization of CD44 was highly reduced under hyperosmotic conditions but poorly affected by membrane cholesterol depletion, revealing that it proceeds mostly via clathrin-coated pits. Finally, we demonstrated that CD44 silencing abolishes RAP-induced tumor cell attachment, revealing that cell surface accumulation of CD44 under LRP-1 blockade is mainly responsible for the stimulation of tumor cell adhesion. Altogether, our data shed light on the LRP-1-mediated internalization of CD44 that appeared critical to define the adhesive properties of tumor cells.
Collapse
|
42
|
Knopfová L, Beneš P, Pekarčíková L, Hermanová M, Masařík M, Pernicová Z, Souček K, Smarda J. c-Myb regulates matrix metalloproteinases 1/9, and cathepsin D: implications for matrix-dependent breast cancer cell invasion and metastasis. Mol Cancer 2012; 11:15. [PMID: 22439866 PMCID: PMC3325857 DOI: 10.1186/1476-4598-11-15] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 03/23/2012] [Indexed: 12/11/2022] Open
Abstract
Background The c-Myb transcription factor is essential for the maintenance of stem-progenitor cells in bone marrow, colon epithelia, and neurogenic niches. c-Myb malfunction contributes to several types of malignancies including breast cancer. However, the function of c-Myb in the metastatic spread of breast tumors remains unexplored. In this study, we report a novel role of c-Myb in the control of specific proteases that regulate the matrix-dependent invasion of breast cancer cells. Results Ectopically expressed c-Myb enhanced migration and ability of human MDA-MB-231 and mouse 4T1 mammary cancer cells to invade Matrigel but not the collagen I matrix in vitro. c-Myb strongly increased the expression/activity of cathepsin D and matrix metalloproteinase (MMP) 9 and significantly downregulated MMP1. The gene coding for cathepsin D was suggested as the c-Myb-responsive gene and downstream effector of the migration-promoting function of c-Myb. Finally, we demonstrated that c-Myb delayed the growth of mammary tumors in BALB/c mice and affected the metastatic potential of breast cancer cells in an organ-specific manner. Conclusions This study identified c-Myb as a matrix-dependent regulator of invasive behavior of breast cancer cells.
Collapse
Affiliation(s)
- Lucia Knopfová
- Department of Experimental Biology, Faculty of Science, Masaryk University, and International Clinical Research Center, CBCE, St. Anne's University Hospital, Brno, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Complex modulation of peptidolytic activity of cathepsin D by sphingolipids. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1811:1097-104. [DOI: 10.1016/j.bbalip.2011.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 08/22/2011] [Accepted: 09/12/2011] [Indexed: 11/18/2022]
|
44
|
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
|
45
|
Enhancement of HLA class II-restricted CD4+ T cell recognition of human melanoma cells following treatment with bryostatin-1. Cell Immunol 2011; 271:392-400. [PMID: 21903207 DOI: 10.1016/j.cellimm.2011.08.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 08/11/2011] [Accepted: 08/12/2011] [Indexed: 12/31/2022]
Abstract
The majority of melanoma cells express detectable levels of HLA class II proteins, and an increased threshold of cell surface class II is crucial for the stimulation of CD4+ T cells. Bryostatin-1, a protein kinase C (PKC) activator, has been considered as a potent chemotherapeutic agent in a variety of in vitro tumor models. Little is known about the role of bryostatin-1 in HLA class II Ag presentation and immune activation in malignant tumors, especially in melanoma. In this study, we show that bryostatin-1 treatment enhances CD4+ T cell recognition of melanoma cells in the context of HLA class II molecules. We also show that bryostatin-1 treatment of melanoma cells increases class II protein levels by upregulating the class II transactivator (CIITA) gene. Flow cytometry and confocal microscopic analyses revealed that bryostatin-1 treatment upregulated the expression of costimulatory molecules (CD80 and CD86) in melanoma cells, which could prolong the interaction of immune cells and tumors. Bryostatin-1 also induced cellular differentiation in melanoma cells, and reduced tumorigenic factors such as pro-cathepsins and matrix-metalloproteinase-9. These data suggest that bryostatin-1 could be used as a chemo-immunotherapeutic agent for reducing tumorigenic potential of melanoma cells while enhancing CD4+ T cell recognition to prevent tumor recurrence.
Collapse
|
46
|
Yelamanchili SV, Chaudhuri AD, Flynn CT, Fox HS. Upregulation of cathepsin D in the caudate nucleus of primates with experimental parkinsonism. Mol Neurodegener 2011; 6:52. [PMID: 21777416 PMCID: PMC3160400 DOI: 10.1186/1750-1326-6-52] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 07/21/2011] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND In Parkinson's disease there is progressive loss of dopamine containing neurons in the substantia nigra pars compacta. The neuronal damage is not limited to the substantia nigra but progresses to other regions of brain, leading to loss of motor control as well as cognitive abnormalities. The purpose of this study was to examine causes of progressive damage in the caudate nucleus, which plays a major role in motor coordination and cognition, in experimental Parkinson's disease. RESULTS Using chronic 1-methyl-4phenyl-1,2,3,6-tetrahydropyridine treatment of rhesus monkeys to model Parkinson's disease, we found a upregulation of Cathepsin D, a lysosomal aspartic protease, in the caudate nucleus of treated monkeys. Immunofluorescence analysis of caudate nucleus brain tissue showed that the number of lysosomes increased concurrently with the increase in Cathepsin D in neurons. In vitro overexpression of Cathepsin D in a human neuroblastoma cell line led to a significant increase in the number of the lysosomes. Such expression also resulted in extralysosomal Cathepsin D and was accompanied by significant neuronal death associated with caspase activation. We examined apoptotic markers and found a strong correlation of Cathepsin D overexpression to apoptosis. CONCLUSIONS Following damage to the substantia nigra resulting in experimental Parkinson's disease, we have identified pathological changes in the caudate nucleus, a likely site of changes leading to the progression of disease. Cathepsin D, implicated in pathogenic mechanisms in other disorders, was increased, and our in vitro studies revealed its overexpression leads to cellular damage and death. This work provides important clues to the progression of Parkinson's, and provides a new target for strategies to ameliorate the progression of this disease.
Collapse
Affiliation(s)
- Sowmya V Yelamanchili
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | | | | | | |
Collapse
|
47
|
Yamamoto K, Kawakubo T, Yasukochi A, Tsukuba T. Emerging roles of cathepsin E in host defense mechanisms. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2011; 1824:105-12. [PMID: 21664991 DOI: 10.1016/j.bbapap.2011.05.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/20/2011] [Accepted: 05/23/2011] [Indexed: 01/07/2023]
Abstract
Cathepsin E is an intracellular aspartic proteinase of the pepsin superfamily, which is predominantly expressed in certain cell types, including the immune system cells and rapidly regenerating gastric mucosal and epidermal keratinocytes. The intracellular localization of this protein varies with different cell types. The endosomal localization is primarily found in antigen-presenting cells and gastric cells. The membrane association is observed with certain cell types such as erythrocytes, osteoclasts, gastric parietal cells and renal proximal tubule cells. This enzyme is also found in the endoplasmic reticulum, Golgi complex and cytosolic compartments in various cell types. In addition to its intracellular localization, cathepsin E occurs in the culture medium of activated phagocytes and cancer cells as the catalytically active enzyme. Its strategic expression and localization thus suggests the association of this enzyme with specific biological functions of the individual cell types. Recent genetic and pharmacological studies have particularly suggested that cathepsin E plays an important role in host defense against cancer cells and invading microorganisms. This review focuses emerging roles of cathepsin E in immune system cells and skin keratinocytes, and in host defense against cancer cells. This article is part of a Special Issue entitled: Proteolysis 50 years after the discovery of lysosome.
Collapse
Affiliation(s)
- Kenji Yamamoto
- Proteolysis Research Laboratory, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| | | | | | | |
Collapse
|
48
|
Basford JE, Wancata L, Hofmann SM, Silva RAGD, Davidson WS, Howles PN, Hui DY. Hepatic deficiency of low density lipoprotein receptor-related protein-1 reduces high density lipoprotein secretion and plasma levels in mice. J Biol Chem 2011; 286:13079-87. [PMID: 21343303 DOI: 10.1074/jbc.m111.229369] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The low density lipoprotein receptor-related protein-1 (LRP1) is known to serve as a chylomicron remnant receptor in the liver responsible for the binding and plasma clearance of apolipoprotein E-containing lipoproteins. Previous in vitro studies have provided evidence to suggest that LRP1 expression may also influence high density lipoprotein (HDL) metabolism. The current study showed that liver-specific LRP1 knock-out (hLrp1(-/-)) mice displayed lower fasting plasma HDL cholesterol levels when compared with hLrp1(+/+) mice. Lecithin:cholesterol acyl transferase and hepatic lipase activities in plasma of hLrp1(-/-) mice were comparable with those observed in hLrp1(+/+) mice, indicating that hepatic LRP1 inactivation does not influence plasma HDL remodeling. Plasma clearance of HDL particles and HDL-associated cholesteryl esters was also similar between hLrp1(+/+) and hLrp1(-/-) mice. In contrast, HDL secretion from primary hepatocytes isolated from hLrp1(-/-) mice was significantly reduced when compared with that observed with hLrp1(+/+) hepatocytes. Biotinylation of cell surface proteins revealed decreased surface localization of the ATP-binding cassette, subfamily A, member 1 (ABCA1) protein, but total cellular ABCA1 level was not changed in hLrp1(-/-) hepatocytes. Finally, hLrp1(-/-) hepatocytes displayed reduced binding capacity for extracellular cathepsin D, resulting in lower intracellular cathepsin D content and impairment of prosaposin activation, a process that is required for membrane translocation of ABCA1 to facilitate cholesterol efflux and HDL secretion. Taken together, these results documented that hepatic LRP1 participates in cellular activation of lysosomal enzymes and through this mechanism, indirectly modulates the production and plasma levels of HDL.
Collapse
Affiliation(s)
- Joshua E Basford
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Masson O, Prébois C, Derocq D, Meulle A, Dray C, Daviaud D, Quilliot D, Valet P, Muller C, Liaudet-Coopman E. Cathepsin-D, a key protease in breast cancer, is up-regulated in obese mouse and human adipose tissue, and controls adipogenesis. PLoS One 2011; 6:e16452. [PMID: 21311773 PMCID: PMC3032791 DOI: 10.1371/journal.pone.0016452] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 12/16/2010] [Indexed: 01/24/2023] Open
Abstract
The aspartic protease cathepsin-D (cath-D) is overexpressed by human epithelial breast cancer cells and is closely correlated with poor prognosis in breast cancer. The adipocyte is one of the most prominent cell types in the tumor-microenvironment of breast cancer, and clinical studies have shown that obesity increases the incidence of breast cancer. Here, we provide the first evidence that cath-D expression is up-regulated in adipose tissue from obese human beings, as well as in adipocytes from the obese C57BI6/J mouse. Cath-D expression is also increased during human and mouse adipocyte differentiation. We show that cath-D silencing in 3T3-F442A murine preadipocytes leads to lipid-depleted cells after adipogenesis induction, and inhibits of the expression of PPARγ, HSL and aP2 adipocyte differentiation markers. Altogether, our findings demonstrate the key role of cath-D in the control of adipogenesis, and suggest that cath-D may be a novel target in obesity.
Collapse
Affiliation(s)
- Olivier Masson
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université Montpellier 1, Montpellier, France
- CRLC Val d'Aurelle Paul Lamarque, Montpellier, France
| | - Christine Prébois
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université Montpellier 1, Montpellier, France
- CRLC Val d'Aurelle Paul Lamarque, Montpellier, France
| | - Danielle Derocq
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université Montpellier 1, Montpellier, France
- CRLC Val d'Aurelle Paul Lamarque, Montpellier, France
| | - Aline Meulle
- Université de Toulouse, UPS, Institut de Médecine Moléculaire de Rangueil, Toulouse, France
- Institute of Pharmacology and Structural Biology CNRS UMR 5089, Toulouse, France
- Université de Toulouse, Toulouse, France
| | - Cédric Dray
- Université de Toulouse, UPS, Institut de Médecine Moléculaire de Rangueil, Toulouse, France
- INSERM, U858, Toulouse, France
| | - Danielle Daviaud
- Université de Toulouse, UPS, Institut de Médecine Moléculaire de Rangueil, Toulouse, France
- INSERM, U858, Toulouse, France
| | - Didier Quilliot
- Service de diabétologie, Maladies métaboliques et nutrition, CHU de Nancy, Nancy, France
| | - Philippe Valet
- Université de Toulouse, UPS, Institut de Médecine Moléculaire de Rangueil, Toulouse, France
- INSERM, U858, Toulouse, France
| | - Catherine Muller
- Institute of Pharmacology and Structural Biology CNRS UMR 5089, Toulouse, France
- Université de Toulouse, Toulouse, France
| | - Emmanuelle Liaudet-Coopman
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université Montpellier 1, Montpellier, France
- CRLC Val d'Aurelle Paul Lamarque, Montpellier, France
- * E-mail:
| |
Collapse
|