1
|
Hämälistö S, Del Valle Batalla F, Yuseff MI, Mattila PK. Endolysosomal vesicles at the center of B cell activation. J Cell Biol 2024; 223:e202307047. [PMID: 38305771 PMCID: PMC10837082 DOI: 10.1083/jcb.202307047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/22/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024] Open
Abstract
The endolysosomal system specializes in degrading cellular components and is crucial to maintaining homeostasis and adapting rapidly to metabolic and environmental cues. Cells of the immune system exploit this network to process antigens or promote cell death by secreting lysosome-related vesicles. In B lymphocytes, lysosomes are harnessed to facilitate the extraction of antigens and to promote their processing into peptides for presentation to T cells, critical steps to mount protective high-affinity antibody responses. Intriguingly, lysosomal vesicles are now considered important signaling units within cells and also display secretory functions by releasing their content to the extracellular space. In this review, we focus on how B cells use pathways involved in the intracellular trafficking, secretion, and function of endolysosomes to promote adaptive immune responses. A basic understanding of such mechanisms poses an interesting frontier for the development of therapeutic strategies in the context of cancer and autoimmune diseases.
Collapse
Affiliation(s)
- Saara Hämälistö
- Institute of Biomedicine, and MediCity Research Laboratories, University of Turku, Turku, Finland
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
- Cancer Research Unit and FICAN West Cancer Centre Laboratory, Turku, Finland
| | - Felipe Del Valle Batalla
- Laboratory of Immune Cell Biology, Department of Cellular and Molecular Biology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María Isabel Yuseff
- Laboratory of Immune Cell Biology, Department of Cellular and Molecular Biology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pieta K. Mattila
- Institute of Biomedicine, and MediCity Research Laboratories, University of Turku, Turku, Finland
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| |
Collapse
|
2
|
Ziegler AR, Dufour A, Scott NE, Edgington-Mitchell LE. Ion Mobility-Based Enrichment-Free N-Terminomics Analysis Reveals Novel Legumain Substrates in Murine Spleen. Mol Cell Proteomics 2024; 23:100714. [PMID: 38199506 PMCID: PMC10862022 DOI: 10.1016/j.mcpro.2024.100714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Aberrant levels of the asparaginyl endopeptidase legumain have been linked to inflammation, neurodegeneration, and cancer, yet our understanding of this protease is incomplete. Systematic attempts to identify legumain substrates have been previously confined to in vitro studies, which fail to mirror physiological conditions and obscure biologically relevant cleavage events. Using high-field asymmetric waveform ion mobility spectrometry (FAIMS), we developed a streamlined approach for proteome and N-terminome analyses without the need for N-termini enrichment. Compared to unfractionated proteomic analysis, we demonstrate FAIMS fractionation improves N-termini identification by >2.5 fold, resulting in the identification of >2882 unique N-termini from limited sample amounts. In murine spleens, this approach identifies 6366 proteins and 2528 unique N-termini, with 235 cleavage events enriched in WT compared to legumain-deficient spleens. Among these, 119 neo-N-termini arose from asparaginyl endopeptidase activities, representing novel putative physiological legumain substrates. The direct cleavage of selected substrates by legumain was confirmed using in vitro assays, providing support for the existence of physiologically relevant extra-lysosomal legumain activity. Combined, these data shed critical light on the functions of legumain and demonstrate the utility of FAIMS as an accessible method to improve depth and quality of N-terminomics studies.
Collapse
Affiliation(s)
- Alexander R Ziegler
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Antoine Dufour
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada
| | - Nichollas E Scott
- Department of Microbiology and Immunology, Peter Doherty Institute, The University of Melbourne, Parkville, Victoria, Australia.
| | - Laura E Edgington-Mitchell
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
3
|
Qian Z, Li B, Meng X, Liao J, Wang G, Li Y, Luo Q, Ye K. Inhibition of asparagine endopeptidase (AEP) effectively treats sporadic Alzheimer's disease in mice. Neuropsychopharmacology 2024; 49:620-630. [PMID: 38030711 PMCID: PMC10789813 DOI: 10.1038/s41386-023-01774-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/12/2023] [Accepted: 11/15/2023] [Indexed: 12/01/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease with cognitive dysfunction as its major clinical symptom. However, there is no disease-modifying small molecular medicine to effectively slow down progression of the disease. Here, we show an optimized asparagine endopeptidase (AEP, also known as δ-secretase) inhibitor, #11 A, that displays an orderly in vivo pharmacokinetics/pharmacodynamics (PK/PD) relationship and robustly attenuates AD pathologies in a sporadic AD mouse model. #11 A is brain permeable with great oral bioavailability. It blocks AEP cleavage of APP and Tau dose-dependently, and significantly decreases Aβ40 and Aβ42 and p-Tau levels in APP/PS1 and Tau P301S mice after oral administration. Notably, #11 A strongly inhibits AEP and prevents mouse APP and Tau fragmentation by AEP, leading to reduction of mouse Aβ42 (mAβ42), mAβ40 and mouse p-Tau181 levels in Thy1-ApoE4/C/EBPβ transgenic mice in a dose-dependent manner. Repeated oral administration of #11 A substantially decreases mAβ aggregation as validated by Aβ PET assay, Tau pathology, neurodegeneration and brain volume reduction, resulting in alleviation of cognitive impairment. Therefore, our results support that #11 A is a disease-modifying preclinical candidate for pharmacologically treating AD.
Collapse
Affiliation(s)
- Zhengjiang Qian
- Faculty of Life and Health Sciences, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Bowei Li
- Shenzhen Institute of Advanced Technology, University of Chinese Academy of Science, Shenzhen, Guangdong, 518055, China
| | - Xin Meng
- Faculty of Life and Health Sciences, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Jianming Liao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Guangxing Wang
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yanjiao Li
- Faculty of Life and Health Sciences, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Qian Luo
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
4
|
Krummenacher D, He W, Kuhn B, Schnider C, Beurier A, Brom V, Sivasothy T, Marty C, Tosstorff A, Hewings DS, Mesch S, Pinard E, Brändlin M, Hochstrasser R, Westwood P, Rothe J, Kronenberger A, Morandi F, Gutbier S, Schuler A, Heer D, Gloria LE, Joedicke L, Rudolph MG, Müller L, Grüninger F, Baumann K, Kaniyappan S, Manevski N, Bartels B. Discovery of Orally Available and Brain Penetrant AEP Inhibitors. J Med Chem 2023; 66:17026-17043. [PMID: 38090813 DOI: 10.1021/acs.jmedchem.3c01804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Alzheimer's Disease (AD) is the most widespread form of dementia, with one of the pathological hallmarks being the formation of neurofibrillary tangles (NFTs). These tangles consist of phosphorylated Tau fragments. Asparagine endopeptidase (AEP) is a key Tau cleaving enzyme that generates aggregation-prone Tau fragments. Inhibition of AEP to reduce the level of toxic Tau fragment formation could represent a promising therapeutic strategy. Here, we report the first orthosteric, selective, orally bioavailable, and brain penetrant inhibitors with an irreversible binding mode. We outline the development of the series starting from reversible molecules and demonstrate the link between inhibition of AEP and reduction of Tau N368 fragment both in vitro and in vivo.
Collapse
Affiliation(s)
- Daniela Krummenacher
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Weiping He
- WuXi AppTec (Wuhan) Co. Ltd., Wuhan East Lake High-Tech Development Zone, 666 GaoXin Road, Wuhan, Hubei 430075, China
| | - Bernd Kuhn
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Christian Schnider
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Angélica Beurier
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Virginie Brom
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Thulase Sivasothy
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Christine Marty
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Andreas Tosstorff
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - David S Hewings
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Stefanie Mesch
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Emmanuel Pinard
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Mathis Brändlin
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Remo Hochstrasser
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Paul Westwood
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Judith Rothe
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Alexandra Kronenberger
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Federica Morandi
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Simon Gutbier
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Angelika Schuler
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Dominik Heer
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Ludivine Esteves Gloria
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Lisa Joedicke
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Markus G Rudolph
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Lutz Müller
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Fiona Grüninger
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Karlheinz Baumann
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Senthilvelrajan Kaniyappan
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Nenad Manevski
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Björn Bartels
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| |
Collapse
|
5
|
Yan L, Han X, Zhang M, Fu Y, Yang F, Li Q, Cheng T. Integrative analysis of TBI data reveals Lgmn as a key player in immune cell-mediated ferroptosis. BMC Genomics 2023; 24:747. [PMID: 38057699 DOI: 10.1186/s12864-023-09842-z] [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: 07/06/2023] [Accepted: 11/24/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a central nervous system disease caused by external trauma, which has complex pathological and physiological mechanisms. The aim of this study was to explore the correlation between immune cell infiltration and ferroptosis post-TBI. METHODS This study utilized the GEO database to download TBI data and performed differentially expressed genes (DEGs) and ferroptosis-related differentially expressed genes (FRDEGs) analysis. DEGs were further analyzed for enrichment using the DAVID 6.8. Immunoinfiltration cell analysis was performed using the ssGSEA package and the Timer2.0 tool. The WGCNA analysis was then used to explore the gene modules in the data set associated with differential expression of immune cell infiltration and to identify the hub genes. The tidyverse package and corrplot package were used to calculate the correlations between hub genes and immune cell infiltration and ferroptosis-marker genes. The miRDB and TargetScan databases were used to predict complementary miRNAs for the Hub genes selected from the WGCNA analysis, and the DIANA-LncBasev3 tool was used to identify target lncRNAs for the miRNAs, constructing an mRNA-miRNA-lncRNA regulatory network. RESULTS A total of 320 DEGs and 21 FRDEGs were identified in GSE128543. GO and KEGG analyses showed that the DEGs after TBI were primarily associated with inflammation and immune response. Xcell and ssGSEA immune infiltration cell analysis showed significant infiltration of T cell CD4+ central memory, T cell CD4+ Th2, B cell memory, B cell naive, monocyte, macrophage, and myeloid dendritic cell activated. The WGCNA analysis identified two modules associated with differentially expressed immune cells and identified Lgmn as a hub gene associated with immune infiltrating cells. Lgmn showed significant correlation with immune cells and ferroptosis-marker genes, including Gpx4, Hspb1, Nfe2l2, Ptgs2, Fth1, and Tfrc. Finally, an mRNA-miRNA-lncRNA regulatory network was constructed using Lgmn. CONCLUSION Our results indicate that there is a certain correlation between ferroptosis and immune infiltrating cells in brain tissue after TBI, and that Lgmn plays an important role in this process.
Collapse
Affiliation(s)
- Liyan Yan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiaonan Han
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Mingkang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yikun Fu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Fei Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Qian Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| | - Tian Cheng
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
6
|
de Lavergne M, Maisonneuve L, Podsypanina K, Manoury B. The role of the antigen processing machinery in the regulation and trafficking of intracellular -Toll-like receptor molecules. Curr Opin Immunol 2023; 84:102375. [PMID: 37562076 DOI: 10.1016/j.coi.2023.102375] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 08/12/2023]
Abstract
Intracellular Toll-like receptors (TLRs) are key components of the innate immune system. Their expression in antigen-presenting cells (APCs), and in particular dendritic cells (DCs), makes them critical in the induction of the adaptive immune response. In DCs, they interact with the chaperone UNC93B1 that mediates their trafficking from the endoplasmic reticulum (ER) to endosomes where they are cleaved by proteases and activated. All these different steps are also shared by major histocompatibility complex class-II (MHCII) molecules. Here, we will discuss the tight relationship intracellular TLRs have with the antigen processing machinery in APCs for their trafficking and activation.
Collapse
Affiliation(s)
- Moïse de Lavergne
- Institut Necker Enfants Malades, INSERM U1151-CNRS UMR 8253, Université Paris Cité, Faculté de Médecine Necker, France
| | - Lucie Maisonneuve
- Institut Necker Enfants Malades, INSERM U1151-CNRS UMR 8253, Université Paris Cité, Faculté de Médecine Necker, France
| | - Katrina Podsypanina
- Institut Necker Enfants Malades, INSERM U1151-CNRS UMR 8253, Université Paris Cité, Faculté de Médecine Necker, France
| | - Bénédicte Manoury
- Institut Necker Enfants Malades, INSERM U1151-CNRS UMR 8253, Université Paris Cité, Faculté de Médecine Necker, France.
| |
Collapse
|
7
|
Solberg R, Lunde NN, Forbord KM, Okla M, Kassem M, Jafari A. The Mammalian Cysteine Protease Legumain in Health and Disease. Int J Mol Sci 2022; 23:ijms232415983. [PMID: 36555634 PMCID: PMC9788469 DOI: 10.3390/ijms232415983] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
The cysteine protease legumain (also known as asparaginyl endopeptidase or δ-secretase) is the only known mammalian asparaginyl endopeptidase and is primarily localized to the endolysosomal system, although it is also found extracellularly as a secreted protein. Legumain is involved in the regulation of diverse biological processes and tissue homeostasis, and in the pathogenesis of various malignant and nonmalignant diseases. In addition to its proteolytic activity that leads to the degradation or activation of different substrates, legumain has also been shown to have a nonproteolytic ligase function. This review summarizes the current knowledge about legumain functions in health and disease, including kidney homeostasis, hematopoietic homeostasis, bone remodeling, cardiovascular and cerebrovascular diseases, fibrosis, aging and senescence, neurodegenerative diseases and cancer. In addition, this review addresses the effects of some marketed drugs on legumain. Expanding our knowledge on legumain will delineate the importance of this enzyme in regulating physiological processes and disease conditions.
Collapse
Affiliation(s)
- Rigmor Solberg
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, N-0316 Oslo, Norway
- Correspondence: (R.S.); (A.J.); Tel.: +47-22-857-514 (R.S.); +45-35-337-423 (A.J.)
| | - Ngoc Nguyen Lunde
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, N-0316 Oslo, Norway
| | - Karl Martin Forbord
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, N-0316 Oslo, Norway
- Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark
| | - Meshail Okla
- Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia
| | - Moustapha Kassem
- Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark
- Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Abbas Jafari
- Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark
- Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Correspondence: (R.S.); (A.J.); Tel.: +47-22-857-514 (R.S.); +45-35-337-423 (A.J.)
| |
Collapse
|
8
|
Sarango G, Richetta C, Pereira M, Kumari A, Ghosh M, Bertrand L, Pionneau C, Le Gall M, Grégoire S, Jeger‐Madiot R, Rosoy E, Subra F, Delelis O, Faure M, Esclatine A, Graff‐Dubois S, Stevanović S, Manoury B, Ramirez BC, Moris A. The Autophagy Receptor TAX1BP1 (T6BP) improves antigen presentation by MHC-II molecules. EMBO Rep 2022; 23:e55470. [PMID: 36215666 PMCID: PMC9724678 DOI: 10.15252/embr.202255470] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 12/12/2022] Open
Abstract
CD4+ T lymphocytes play a major role in the establishment and maintenance of immunity. They are activated by antigenic peptides derived from extracellular or newly synthesized (endogenous) proteins presented by the MHC-II molecules. The pathways leading to endogenous MHC-II presentation remain poorly characterized. We demonstrate here that the autophagy receptor, T6BP, influences both autophagy-dependent and -independent endogenous presentation of HIV- and HCMV-derived peptides. By studying the immunopeptidome of MHC-II molecules, we show that T6BP affects both the quantity and quality of peptides presented. T6BP silencing induces the mislocalization of the MHC-II-loading compartments and rapid degradation of the invariant chain (CD74) without altering the expression and internalization kinetics of MHC-II molecules. Defining the interactome of T6BP, we identify calnexin as a T6BP partner. We show that the calnexin cytosolic tail is required for this interaction. Remarkably, calnexin silencing replicates the functional consequences of T6BP silencing: decreased CD4+ T cell activation and exacerbated CD74 degradation. Altogether, we unravel T6BP as a key player of the MHC-II-restricted endogenous presentation pathway, and we propose one potential mechanism of action.
Collapse
Affiliation(s)
- Gabriela Sarango
- Université Paris‐Saclay, CEA, CNRSInstitute for Integrative Biology of the Cell (I2BC)Gif‐sur‐YvetteFrance,Sorbonne UniversitéINSERM, CNRS, Center for Immunology and Microbial Infections (CIMI‐Paris)ParisFrance
| | - Clémence Richetta
- Sorbonne UniversitéINSERM, CNRS, Center for Immunology and Microbial Infections (CIMI‐Paris)ParisFrance,LBPA, ENS‐Paris Saclay, CNRS UMR8113Université Paris SaclayGif‐sur‐YvetteFrance
| | - Mathias Pereira
- Université Paris‐Saclay, CEA, CNRSInstitute for Integrative Biology of the Cell (I2BC)Gif‐sur‐YvetteFrance,Sorbonne UniversitéINSERM, CNRS, Center for Immunology and Microbial Infections (CIMI‐Paris)ParisFrance
| | - Anita Kumari
- Université Paris‐Saclay, CEA, CNRSInstitute for Integrative Biology of the Cell (I2BC)Gif‐sur‐YvetteFrance,Sorbonne UniversitéINSERM, CNRS, Center for Immunology and Microbial Infections (CIMI‐Paris)ParisFrance
| | - Michael Ghosh
- Department of Immunology, Institute for Cell BiologyUniversity of TübingenTübingenGermany
| | - Lisa Bertrand
- Université Paris‐Saclay, CEA, CNRSInstitute for Integrative Biology of the Cell (I2BC)Gif‐sur‐YvetteFrance,Sorbonne UniversitéINSERM, CNRS, Center for Immunology and Microbial Infections (CIMI‐Paris)ParisFrance
| | - Cédric Pionneau
- Sorbonne UniversitéINSERM, UMS Production et Analyse de Données en Sciences de la vie et en Santé, PASS, Plateforme Post‐génomique de la Pitié SalpêtrièreParisFrance
| | - Morgane Le Gall
- 3P5 proteom'IC facilityUniversité de Paris, Institut Cochin, INSERM U1016, CNRS‐UMR 8104ParisFrance
| | - Sylvie Grégoire
- Université Paris‐Saclay, CEA, CNRSInstitute for Integrative Biology of the Cell (I2BC)Gif‐sur‐YvetteFrance,Sorbonne UniversitéINSERM, CNRS, Center for Immunology and Microbial Infections (CIMI‐Paris)ParisFrance
| | - Raphaël Jeger‐Madiot
- Sorbonne UniversitéINSERM, CNRS, Center for Immunology and Microbial Infections (CIMI‐Paris)ParisFrance,Present address:
Sorbonne Université, INSERM U959, Immunology‐Immunopathology‐Immunotherapy (i3)ParisFrance
| | - Elina Rosoy
- Sorbonne UniversitéINSERM, CNRS, Center for Immunology and Microbial Infections (CIMI‐Paris)ParisFrance
| | - Frédéric Subra
- LBPA, ENS‐Paris Saclay, CNRS UMR8113Université Paris SaclayGif‐sur‐YvetteFrance
| | - Olivier Delelis
- LBPA, ENS‐Paris Saclay, CNRS UMR8113Université Paris SaclayGif‐sur‐YvetteFrance
| | - Mathias Faure
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de LyonLyonFrance,Equipe Labellisée par la Fondation pour la Recherche Médicale, FRM
| | - Audrey Esclatine
- Université Paris‐Saclay, CEA, CNRSInstitute for Integrative Biology of the Cell (I2BC)Gif‐sur‐YvetteFrance
| | - Stéphanie Graff‐Dubois
- Sorbonne UniversitéINSERM, CNRS, Center for Immunology and Microbial Infections (CIMI‐Paris)ParisFrance,Present address:
Sorbonne Université, INSERM U959, Immunology‐Immunopathology‐Immunotherapy (i3)ParisFrance
| | - Stefan Stevanović
- Department of Immunology, Institute for Cell BiologyUniversity of TübingenTübingenGermany
| | - Bénédicte Manoury
- Institut Necker Enfants Malades, INSERM U1151‐CNRS UMR 8253, Faculté de médecine NeckerUniversité de ParisParisFrance
| | - Bertha Cecilia Ramirez
- Université Paris‐Saclay, CEA, CNRSInstitute for Integrative Biology of the Cell (I2BC)Gif‐sur‐YvetteFrance,Sorbonne UniversitéINSERM, CNRS, Center for Immunology and Microbial Infections (CIMI‐Paris)ParisFrance
| | - Arnaud Moris
- Université Paris‐Saclay, CEA, CNRSInstitute for Integrative Biology of the Cell (I2BC)Gif‐sur‐YvetteFrance,Sorbonne UniversitéINSERM, CNRS, Center for Immunology and Microbial Infections (CIMI‐Paris)ParisFrance
| |
Collapse
|
9
|
Kawasaki T, Ikegawa M, Kawai T. Antigen Presentation in the Lung. Front Immunol 2022; 13:860915. [PMID: 35615351 PMCID: PMC9124800 DOI: 10.3389/fimmu.2022.860915] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/15/2022] [Indexed: 12/28/2022] Open
Abstract
The lungs are constantly exposed to environmental and infectious agents such as dust, viruses, fungi, and bacteria that invade the lungs upon breathing. The lungs are equipped with an immune defense mechanism that involves a wide variety of immunological cells to eliminate these agents. Various types of dendritic cells (DCs) and macrophages (MACs) function as professional antigen-presenting cells (APCs) that engulf pathogens through endocytosis or phagocytosis and degrade proteins derived from them into peptide fragments. During this process, DCs and MACs present the peptides on their major histocompatibility complex class I (MHC-I) or MHC-II protein complex to naïve CD8+ or CD4+ T cells, respectively. In addition to these cells, recent evidence supports that antigen-specific effector and memory T cells are activated by other lung cells such as endothelial cells, epithelial cells, and monocytes through antigen presentation. In this review, we summarize the molecular mechanisms of antigen presentation by APCs in the lungs and their contribution to immune response.
Collapse
Affiliation(s)
| | | | - Taro Kawai
- *Correspondence: Takumi Kawasaki, ; Taro Kawai,
| |
Collapse
|
10
|
Wang S, Wang S, Wang X, Xu Y, Zhang X, Han Y, Yan H, Liu L, Wang L, Ye H, Li X. Effects of Icariin on Modulating Gut Microbiota and Regulating Metabolite Alterations to Prevent Bone Loss in Ovariectomized Rat Model. Front Endocrinol (Lausanne) 2022; 13:874849. [PMID: 35399950 PMCID: PMC8988140 DOI: 10.3389/fendo.2022.874849] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Postmenopausal osteoporosis (PMOP) is an estrogen deficiency-induced bone loss, which has been shown an association with an altered gut microbiota (GM). Gut microbiota-bone axis has been recognized as a crucial mediator for bone homeostasis. Icariin (ICA) is an effective agent to delay bone loss by regulating the bone homeostasis. Thus, we hypothesize that ICA can prevent bone loss by modulating GM and regulating metabolite alterations. The effects of ICA on bone metabolism improvement in ovariectomized (OVX) rats and their relationships with the GM and fecal metabolites were investigated. Micro-computed tomography (micro-CT) and hematoxylin-eosin (HE) staining showed a typical bone boss in OVX group, while ICA or estradiol (E2) administration exhibited positive effects on bone micro-architecture improvement. The GM such as Actinobacteria, Gammaproteobacteria, Erysipelotrichi, Erysipelotrichales, Enterobacteriales, Actinomycetales, Ruminococcus and Oscillospira significantly correlated to serum bone Gla-protein (BGP), receptor activator of nuclear factor-κB (RANK), receptor activator of nuclear factor-κB ligand (RANKL), osteoprotegerin (OPG) and tartrate resistant acid phosphatase (TRACP). Further t-test revealed a substantial variation of the GM and fecal metabolites in different treatments. Among them, Lachnoclostridium, Butyricimonas, Rikenella, Paraprevolla, Adlercreutzia, Enterorhabdus, Anaerovorax, Allobaculum, Elusimicrobium, Lactococcus, Globicatella and Lactobacillus were probably the key microbial communities driving the change of bile acid, amino acid and fatty acid, thereby leading to an improvement of PMOP. The significant up-regulation of L-Saccharopine, 1-Aminocyclohexadieneacid and linoleic acid after ICA administration suggested important contributions of amino acid and fatty acid metabolisms in the prevention and treatment of PMOP. Taken together, our study has provided new perspectives to better understand the effects of ICA on PMOP improvement by regulating GM and the associated fecal metabolites. Our findings contribute to develop ICA as a potential therapy for PMOP.
Collapse
Affiliation(s)
- Shanshan Wang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Shengjie Wang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiaoning Wang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Key Laboratory of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yunteng Xu
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xin Zhang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yidan Han
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hui Yan
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Basic Discipline Laboratory of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Linglong Liu
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Basic Discipline Laboratory of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lili Wang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hongzhi Ye
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xihai Li
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Key Laboratory of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
11
|
Watts C. Lysosomes and lysosome‐related organelles in immune responses. FEBS Open Bio 2022; 12:678-693. [PMID: 35220694 PMCID: PMC8972042 DOI: 10.1002/2211-5463.13388] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/25/2022] [Indexed: 11/17/2022] Open
Abstract
The catabolic, degradative capacity of the endo‐lysosome system is put to good use in mammalian immune responses as is their recently established status as signaling platforms. From the ‘creative destruction’ of antigenic and ‘self’ material for antigen presentation to T cells to the re‐purposing of lysosomes as toxic exocytosable lysosome‐related organelles (granules) in leukocytes such as CD8 T cells and eosinophils, endo‐lysosomes are key players in host defense. Signaled responses to some pathogen products initiate in endo‐lysosomes and these organelles are emerging as important in distinct ways in the unique immunobiology of dendritic cells. Potential self‐inflicted toxicity from lysosomal and granule proteases is countered by expression of serpin and cystatin family members.
Collapse
Affiliation(s)
- Colin Watts
- Division of Cell Signalling & Immunology School of Life Sciences University of Dundee Dundee DD1 5EH UK
| |
Collapse
|
12
|
Amarnath S, Brown ML. Harnessing proteases for T regulatory cell immunotherapy. Eur J Immunol 2020; 50:770-778. [PMID: 32383480 DOI: 10.1002/eji.201948270] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 03/08/2020] [Accepted: 05/06/2020] [Indexed: 12/27/2022]
Abstract
The immune system is tightly regulated by a subset of T cells defined as regulatory T cells (Tregs). Tregs maintain immune homeostasis by restraining unwarranted immune cell activation and effector function. Here, we discuss an important but underappreciated role of proteases in controlling Treg function. Proteases regulate a number of vital processes that determine T cell immune responses and some of them such as furin, ADAM (through regulating LAG receptor), MALT, and asparaginyl endopeptidase are implicated in Treg immunobiology. Targeted protease inhibition, using either small molecule inhibitors or gene deficient mice has demonstrated their specificity in modulating Treg function in experimental murine models. These data further highlight the ability of proteases to specifically regulate Tregs but no other T effector lineages. Taken together, it is apparent that incorporating proteases as targets within Treg cell engineering protocols may enable generation of robust Treg cellular therapeutics. These engineered Tregs may possess enhanced regulatory function along with resistance to lineage deviation in inflammatory disease such as colitis and graft versus host disease. Within this review, we summarize research on the role of proteases in regulating Treg function and discuss the translational potential of harnessing Treg function by targeting protease driven regulatory pathways.
Collapse
Affiliation(s)
- Shoba Amarnath
- NUTranslational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Marnie L Brown
- NUTranslational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
13
|
Tohme M, Maisonneuve L, Achour K, Dussiot M, Maschalidi S, Manoury B. TLR7 trafficking and signaling in B cells is regulated by the MHCII-associated invariant chain. J Cell Sci 2020; 133:jcs.236711. [PMID: 32079661 DOI: 10.1242/jcs.236711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 02/04/2020] [Indexed: 11/20/2022] Open
Abstract
Toll-like receptor 7 (TLR7) is an endosomal receptor that recognizes single-stranded RNA from viruses. Its trafficking and activation is regulated by the endoplasmic reticulum (ER) chaperone UNC93B1 and lysosomal proteases. UNC93B1 also modulates major histocompatibility complex class II (MHCII) antigen presentation, and deficiency in MHCII protein diminishes TLR9 signaling. These results indicate a link between proteins that regulate both innate and adaptive responses. Here, we report that TLR7 resides in lysosomes and interacts with the MHCII-chaperone molecule, the invariant chain (Ii) or CD74, in B cells. In the absence of CD74, TLR7 displays both ER and lysosomal localization, leading to an increase in pro-inflammatory cytokine production. Furthermore, stimulation with TLR7 but not TLR9, is inefficient in boosting antigen presentation in Ii-deficient cells. In contrast, in B cells lacking TLR7 or mutated for UNC93B1, which are able to trigger TLR7 activation, antigen presentation is enhanced. This suggests that TLR7 signaling in B cells is controlled by the Ii chain.
Collapse
Affiliation(s)
- Mira Tohme
- Nkarta Therapeutics, South San Fransisco, CA 94080, USA
| | - Lucie Maisonneuve
- Institut Necker Enfant Malade, INSERM U1151-CNRS UMR 8253, 75015 Paris, France.,Université de Paris, Faculté de médecine, 75015 Paris, France
| | - Karim Achour
- Institut de recherche Servier, 3 rue de la république, 92150 Suresnes, France
| | - Michaël Dussiot
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, 75015 Paris, France
| | - Sophia Maschalidi
- VIB-UGent Center for Inflammation Research, UGent-VIB Research Building FSVM, Technologiepark 71, 9052 Ghent, Belgium
| | - Bénédicte Manoury
- Institut Necker Enfant Malade, INSERM U1151-CNRS UMR 8253, 75015 Paris, France .,Université de Paris, Faculté de médecine, 75015 Paris, France
| |
Collapse
|
14
|
Zhang Z, Tian Y, Ye K. δ-secretase in neurodegenerative diseases: mechanisms, regulators and therapeutic opportunities. Transl Neurodegener 2020; 9:1. [PMID: 31911834 PMCID: PMC6943888 DOI: 10.1186/s40035-019-0179-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/26/2019] [Indexed: 11/10/2022] Open
Abstract
Mammalian asparagine endopeptidase (AEP) is a cysteine protease that cleaves its protein substrates on the C-terminal side of asparagine residues. Converging lines of evidence indicate that AEP may be involved in the pathogenesis of several neurological diseases, including Alzheimer's disease, Parkinson's disease, and frontotemporal dementia. AEP is activated in the aging brain, cleaves amyloid precursor protein (APP) and promotes the production of amyloid-β (Aβ). We renamed AEP to δ-secretase to emphasize its role in APP fragmentation and Aβ production. AEP also cleaves other substrates, such as tau, α-synuclein, SET, and TAR DNA-binding protein 43, generating neurotoxic fragments and disturbing their physiological functions. The activity of δ-secretase is tightly regulated at both the transcriptional and posttranslational levels. Here, we review the recent advances in the role of δ-secretase in neurodegenerative diseases, with a focus on its biochemical properties and the transcriptional and posttranslational regulation of its activity, and discuss the clinical implications of δ-secretase as a diagnostic biomarker and therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060 People’s Republic of China
| | - Ye Tian
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060 People’s Republic of China
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322 USA
| |
Collapse
|
15
|
Anderson BM, de Almeida LGN, Sekhon H, Young D, Dufour A, Edgington-Mitchell LE. N-Terminomics/TAILS Profiling of Macrophages after Chemical Inhibition of Legumain. Biochemistry 2019; 59:329-340. [PMID: 31774660 DOI: 10.1021/acs.biochem.9b00821] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Legumain (asparaginyl endopeptidase) is the only protease with a preference for cleavage after asparagine residues. Increased legumain activity is a hallmark of inflammation, neurodegenerative diseases, and cancer, and legumain inhibitors have exhibited therapeutic effects in mouse models of these pathologies. Improved knowledge of its substrates and cellular functions is a requisite to further validation of legumain as a drug target. We, therefore, aimed to investigate the effects of legumain inhibition in macrophages using an unbiased and systematic approach. By shotgun proteomics, we identified 16 094 unique peptides in RAW264.7 cells. Among these, 326 unique peptides were upregulated in response to legumain inhibition, while 241 were downregulated. Many of these proteins were associated with mitochondria and metabolism, especially iron metabolism, indicating that legumain may have a previously unknown impact on related processes. Furthermore, we used N-terminomics/TAILS (terminal amine isotopic labeling of substrates) to identify potential substrates of legumain. We identified three new proteins that are cleaved after asparagine residues, which may reflect legumain-dependent cleavage. We confirmed that frataxin, a mitochondrial protein associated with the formation of iron-sulfur clusters, can be cleaved by legumain. This further asserts a potential contribution of legumain to mitochondrial function and iron metabolism. Lastly, we also identified a potential new cleavage site within legumain itself that may give rise to a 25 kDa form of legumain that has previously been observed in multiple cell and tissue types. Collectively, these data shed new light on the potential functions of legumain and will be critical for understanding its contribution to disease.
Collapse
Affiliation(s)
- Bethany M Anderson
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Parkville , Victoria 3052 , Australia
| | - Luiz G N de Almeida
- Department of Physiology and Pharmacology , University of Calgary , Calgary , Alberta T2N 4N1 , Canada.,McCaig Institute for Bone and Joint Health , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
| | - Henna Sekhon
- Department of Physiology and Pharmacology , University of Calgary , Calgary , Alberta T2N 4N1 , Canada.,McCaig Institute for Bone and Joint Health , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
| | - Daniel Young
- Department of Physiology and Pharmacology , University of Calgary , Calgary , Alberta T2N 4N1 , Canada.,McCaig Institute for Bone and Joint Health , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
| | - Antoine Dufour
- Department of Physiology and Pharmacology , University of Calgary , Calgary , Alberta T2N 4N1 , Canada.,McCaig Institute for Bone and Joint Health , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
| | - Laura E Edgington-Mitchell
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Parkville , Victoria 3052 , Australia.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences , Monash University , Parkville , Victoria 3052 , Australia.,Department of Oral and Maxillofacial Surgery , New York University College of Dentistry, Bluestone Center for Clinical Research , New York , New York 10010 , United States
| |
Collapse
|
16
|
Eddie SL, Gregson A, Graham E, Burton S, Harrison T, Burden R, Scott CJ, Mullan PB, Williams R. Identification and SAR exploration of a novel series of Legumain inhibitors. Bioorg Med Chem Lett 2019; 29:1546-1548. [DOI: 10.1016/j.bmcl.2019.03.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/25/2019] [Accepted: 03/15/2019] [Indexed: 10/27/2022]
|
17
|
Chen X, Wang C, Liao K, Zhou S, Cao L, Chen J, Xu C, Lin Y. USP17 Suppresses Tumorigenesis and Tumor Growth through Deubiquitinating AEP. Int J Biol Sci 2019; 15:738-748. [PMID: 30906206 PMCID: PMC6429017 DOI: 10.7150/ijbs.30106] [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: 09/21/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022] Open
Abstract
Ubiquitin-specific protease 17 (USP17), a novel member of deubiquitinase, is reported to play essential roles in several solid tumors. However, the expression and function of USP17 in breast cancer tumorigenesis remains ambiguity. Here we found that the mRNA level of USP17 was lower in breast cancer tissues than normal tissues. Meanwhile, higher USP17 level was detected in normal epithelial cell MCF-10A and a less-malignant cell MCF-7 than malignant cell line MDA-MB-231. Inhibition of USP17 in MCF7 cells enhanced tumorigenesis and tumor growth while overexpression of USP17 in malignant MDA-MB-231 cells reduced its tumorigenesis and growth ability in vitro and in vivo. Further study revealed that USP17 interacted with and deubiquitinated Asparaginyl endopeptidase (AEP), resulting in decreased protein levels of AEP. Moreover, knockdown of AEP inhibited breast cancer tumorigenesis and growth in vitro and in vivo through the inactivation of ERK signaling. Taken together, our works indicate that USP17 deubiquitinates AEP, down-regulates its protein level, and inhibits breast cancer tumorigenesis through disturbing ERK signaling. Thus, our data suggests that USP17 is a potential tumor suppressor in breast cancer and AEP is a promising target in breast cancer therapy.
Collapse
Affiliation(s)
- Xi Chen
- CAS key laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Chen Wang
- Shanghai Institute of Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Keman Liao
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao-tong University, Shanghai, 200127, China
| | - Sunhai Zhou
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao-tong University, Shanghai, 200127, China
| | - Lu Cao
- Department of Radiation Oncology, Ruijin Hospital, School of Medicine, Shanghai Jiao-tong University, Shanghai 200025, China
| | - Jiayi Chen
- Department of Radiation Oncology, Ruijin Hospital, School of Medicine, Shanghai Jiao-tong University, Shanghai 200025, China
| | - Cheng Xu
- Department of Radiation Oncology, Ruijin Hospital, School of Medicine, Shanghai Jiao-tong University, Shanghai 200025, China
| | - Yingying Lin
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao-tong University, Shanghai, 200127, China
| |
Collapse
|
18
|
Thibodeau J, Moulefera MA, Balthazard R. On the structure–function of MHC class II molecules and how single amino acid polymorphisms could alter intracellular trafficking. Hum Immunol 2019; 80:15-31. [DOI: 10.1016/j.humimm.2018.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/25/2018] [Accepted: 10/01/2018] [Indexed: 12/01/2022]
|
19
|
Freeley S, Cardone J, Günther SC, West EE, Reinheckel T, Watts C, Kemper C, Kolev MV. Asparaginyl Endopeptidase (Legumain) Supports Human Th1 Induction via Cathepsin L-Mediated Intracellular C3 Activation. Front Immunol 2018; 9:2449. [PMID: 30405635 PMCID: PMC6207624 DOI: 10.3389/fimmu.2018.02449] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 10/04/2018] [Indexed: 12/31/2022] Open
Abstract
Autocrine activation of the complement receptors C3aR and CD46 by complement activation components C3a and C3b produced through C3 cleavage by the protease cathepsin L (CTSL) during T cell stimulation is a requirement for IFN-γ production and Th1 induction in human CD4+ T cells. Thus, lack of autocrine CD46 activation, such as in CD46-deficient patients, is associated with defective Th1 responses and recurrent infections. We have identified LGMN [the gene coding for legumain, also known as asparaginyl endopeptidase (AEP)] as one of the key genes induced by CD46 co-stimulation during human CD4+ T cell activation. AEP processes and activates a range of proteins, among those α1-thymosin and CTSL, which both drive intrinsically Th1 activity-but has so far not been described to be functionally active in human T cells. Here we found that pharmacological inhibition of AEP during activation of human CD4+ T cells reduced CTSL activation and the CTSL-mediated generation of intracellular C3a. This translated into a specific reduction of IFN-γ production without affecting cell proliferation or survival. In line with these findings, CD4+ T cells isolated from Lgmn -/- mice also displayed a specific defect in IFN-γ secretion and Th1 induction. Furthermore, we did not observe a role for AEP-driven autocrine α1-thymosin activation in T cell-derived IFN-γ production. These data suggest that AEP is an "upstream" activator of the CTSL-C3-IFN-γ axis in human CD4+ T cells and hence an important supporter of human Th1 induction.
Collapse
Affiliation(s)
- Simon Freeley
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - John Cardone
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Sira C Günther
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom.,Institut für Medizinische Virologie, University of Zurich, Zurich, Switzerland
| | - Erin E West
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Thomas Reinheckel
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs University Freiburg, and German Cancer Consortium (DKTK), Freiburg, Germany
| | - Colin Watts
- Division of Cell Signaling & Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Claudia Kemper
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom.,Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, United States.,Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Martin V Kolev
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| |
Collapse
|
20
|
Ilgová J, Jedličková L, Dvořáková H, Benovics M, Mikeš L, Janda L, Vorel J, Roudnický P, Potěšil D, Zdráhal Z, Gelnar M, Kašný M. A novel type I cystatin of parasite origin with atypical legumain-binding domain. Sci Rep 2017; 7:17526. [PMID: 29235483 PMCID: PMC5727476 DOI: 10.1038/s41598-017-17598-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 11/28/2017] [Indexed: 12/26/2022] Open
Abstract
Parasite inhibitors of cysteine peptidases are known to influence a vast range of processes linked to a degradation of either the parasites' own proteins or proteins native to their hosts. We characterise a novel type I cystatin (stefin) found in a sanguinivorous fish parasite Eudiplozoon nipponicum (Platyhelminthes: Monogenea). We have identified a transcript of its coding gene in the transcriptome of adult worms. Its amino acid sequence is similar to other stefins except for containing a legumain-binding domain, which is in this type of cystatins rather unusual. As expected, the recombinant form of E. nipponicum stefin (rEnStef) produced in Escherichia coli inhibits clan CA peptidases - cathepsins L and B of the worm - via the standard papain-binding domain. It also blocks haemoglobinolysis by cysteine peptidases in the worm's excretory-secretory products and soluble extracts. Furthermore, we had confirmed its ability to inhibit clan CD asparaginyl endopeptidase (legumain). The presence of a native EnStef in the excretory-secretory products of adult worms, detected by mass spectrometry, suggests that this protein has an important biological function at the host-parasite interface. We discuss the inhibitor's possible role in the regulation of blood digestion, modulation of antigen presentation, and in the regeneration of host tissues.
Collapse
Affiliation(s)
- Jana Ilgová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Brno, 611 37, Czech Republic.
| | - Lucie Jedličková
- Department of Parasitology, Faculty of Science, Charles University, Prague, 128 44, Czech Republic
| | - Hana Dvořáková
- Department of Parasitology, Faculty of Science, Charles University, Prague, 128 44, Czech Republic
| | - Michal Benovics
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Brno, 611 37, Czech Republic
| | - Libor Mikeš
- Department of Parasitology, Faculty of Science, Charles University, Prague, 128 44, Czech Republic
| | - Lubomír Janda
- Central European Institute of Technology, Masaryk University, Brno, 625 00, Czech Republic
| | - Jiří Vorel
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Brno, 611 37, Czech Republic
| | - Pavel Roudnický
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Brno, 611 37, Czech Republic
| | - David Potěšil
- Central European Institute of Technology, Masaryk University, Brno, 625 00, Czech Republic
| | - Zbyněk Zdráhal
- Central European Institute of Technology, Masaryk University, Brno, 625 00, Czech Republic
| | - Milan Gelnar
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Brno, 611 37, Czech Republic
| | - Martin Kašný
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Brno, 611 37, Czech Republic.,Department of Parasitology, Faculty of Science, Charles University, Prague, 128 44, Czech Republic
| |
Collapse
|
21
|
Abstract
Unlike B cells, CD8-positive and CD4-positive T cells of the adaptive immune system do not recognize intact foreign proteins but instead recognize polypeptide fragments of potential antigens. These antigenic peptides are expressed on the surface of antigen presenting cells bound to MHC class I and MHC class II proteins. Here, we review the basics of antigen acquisition by antigen presenting cells, antigen proteolysis into polypeptide fragments, antigenic peptide binding to MHC proteins, and surface display of both MHC class I-peptide and MHC class II-peptide complexes.
Collapse
|
22
|
Rawlings ND. Using the MEROPS Database for Investigation of Lysosomal Peptidases, Their Inhibitors, and Substrates. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2017; 1594:213-226. [PMID: 28456986 DOI: 10.1007/978-1-4939-6934-0_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
This chapter describes how to retrieve data on lysosomal peptidases from the MEROPS database for proteolytic enzymes, their substrates and inhibitors ( http://merops.sanger.ac.uk ). Features described in this chapter include the summary page, pages for structure, interactions with inhibitors, substrates, literature and involvement in physiological pathways, and how to download data from the MEROPS FTP site. The lysosomal peptidase legumain is used as an example.
Collapse
Affiliation(s)
- Neil D Rawlings
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK.
| |
Collapse
|
23
|
Adler LN, Jiang W, Bhamidipati K, Millican M, Macaubas C, Hung SC, Mellins ED. The Other Function: Class II-Restricted Antigen Presentation by B Cells. Front Immunol 2017; 8:319. [PMID: 28386257 PMCID: PMC5362600 DOI: 10.3389/fimmu.2017.00319] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/07/2017] [Indexed: 12/31/2022] Open
Abstract
Mature B lymphocytes (B cells) recognize antigens using their B cell receptor (BCR) and are activated to become antibody-producing cells. In addition, and integral to the development of a high-affinity antibodies, B cells utilize the specialized major histocompatibility complex class II (MHCII) antigen presentation pathway to process BCR-bound and internalized protein antigens and present selected peptides in complex with MHCII to CD4+ T cells. This interaction influences the fate of both types of lymphocytes and shapes immune outcomes. Specific, effective, and optimally timed antigen presentation by B cells requires well-controlled intracellular machinery, often regulated by the combined effects of several molecular events. Here, we delineate and summarize these events in four steps along the antigen presentation pathway: (1) antigen capture and uptake by B cells; (2) intersection of internalized antigen/BCRs complexes with MHCII in peptide-loading compartments; (3) generation and regulation of MHCII/peptide complexes; and (4) exocytic transport for presentation of MHCII/peptide complexes at the surface of B cells. Finally, we discuss modulation of the MHCII presentation pathway across B cell development and maturation to effector cells, with an emphasis on the shaping of the MHCII/peptide repertoire by two key antigen presentation regulators in B cells: HLA-DM and HLA-DO.
Collapse
Affiliation(s)
- Lital N Adler
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Program in Immunology, Stanford University, Stanford, CA, USA
| | - Wei Jiang
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Program in Immunology, Stanford University, Stanford, CA, USA
| | | | | | - Claudia Macaubas
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Program in Immunology, Stanford University, Stanford, CA, USA
| | - Shu-Chen Hung
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Program in Immunology, Stanford University, Stanford, CA, USA
| | - Elizabeth D Mellins
- Department of Pediatrics, Stanford University, Stanford, CA, USA; Program in Immunology, Stanford University, Stanford, CA, USA
| |
Collapse
|
24
|
Counter Selection Substrate Library Strategy for Developing Specific Protease Substrates and Probes. Cell Chem Biol 2016; 23:1023-35. [PMID: 27478158 DOI: 10.1016/j.chembiol.2016.05.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/11/2016] [Accepted: 05/30/2016] [Indexed: 01/29/2023]
Abstract
Legumain (AEP) is a lysosomal cysteine protease that was first characterized in leguminous seeds and later discovered in higher eukaryotes. AEP upregulation is linked to a number of diseases including inflammation, arteriosclerosis, and tumorigenesis. Thus this protease is an excellent molecular target for the development of new chemical markers. We deployed a hybrid combinatorial substrate library (HyCoSuL) approach to obtain P1-Asp fluorogenic substrates and biotin-labeled inhibitors that targeted legumain. Since this approach led to probes that were also recognized by caspases, we introduced a Counter Selection Substrate Library (CoSeSuL) approach that biases the peptidic scaffold against caspases, thus delivering highly selective legumain probes. The selectivity of these tools was validated using M38L and HEK293 cells. We also propose that the CoSeSuL methodology can be considered as a general principle in the design of selective probes for other protease families where selectivity is difficult to achieve by conventional sequence-based profiling.
Collapse
|
25
|
Zhang Z, Xie M, Ye K. Asparagine endopeptidase is an innovative therapeutic target for neurodegenerative diseases. Expert Opin Ther Targets 2016; 20:1237-45. [PMID: 27115710 DOI: 10.1080/14728222.2016.1182990] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Asparagine endopeptidase (AEP) is a pH-dependent endolysosomal cysteine protease that cleaves its substrates after asparagine residues. Our most recent study identifies that it possesses the delta-secretase activity, and that it is implicated in numerous neurological diseases such as Alzheimer's disease (AD) and stroke. Accumulating evidence supports that the inhibition of AEP exhibits beneficial effects for treating these devastating diseases. AREAS COVERED Based on recent evidence, it is clear that AEP cleaves its substrate, such as amyloid precursor protein (APP), tau and SET, and plays a critical role in neuronal cell death in various neurodegenerative diseases and stroke. In this article, the basic biology of AEP, its knockout phenotypes in mouse models, its substrates in neurodegenerative diseases, and its small peptidyl inhibitors and prodrugs are discussed. In addition, we discuss the potential of AEP as a novel therapeutic target for neurodegenerative diseases. EXPERT OPINION AEP plays a unique role in numerous biological processes, depending on both pH and context. Most striking is our most recent finding; that AEP is activated in an age-dependent manner and simultaneously cleaves both APP and tau, thereby unifying both major pathological events in AD. Thus, AEP acts as an innovative trigger for neurodegenerative diseases. Inhibition of AEP will provide a disease-modifying treatment for neurodegenerative diseases including AD.
Collapse
Affiliation(s)
- Zhentao Zhang
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China.,b Department of Pathology and Laboratory Medicine , Emory University School of Medicine , Atlanta , GA , USA
| | - Manling Xie
- b Department of Pathology and Laboratory Medicine , Emory University School of Medicine , Atlanta , GA , USA
| | - Keqiang Ye
- b Department of Pathology and Laboratory Medicine , Emory University School of Medicine , Atlanta , GA , USA
| |
Collapse
|
26
|
Flat SAR of P3-methylsulphonamide based small molecule legumain inhibitors. Bioorg Med Chem Lett 2016; 26:413-416. [DOI: 10.1016/j.bmcl.2015.11.101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 11/26/2015] [Accepted: 11/27/2015] [Indexed: 11/17/2022]
|
27
|
Ness KA, Eddie SL, Higgins CA, Templeman A, D'Costa Z, Gaddale KKD, Bouzzaoui S, Jordan L, Janssen D, Harrison T, Burkamp F, Young A, Burden R, Scott CJ, Mullan PB, Williams R. Development of a potent and selective cell penetrant Legumain inhibitor. Bioorg Med Chem Lett 2015; 25:5642-5. [PMID: 26522952 DOI: 10.1016/j.bmcl.2015.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 09/30/2015] [Accepted: 10/01/2015] [Indexed: 11/27/2022]
Abstract
This Letter describes the continued SAR exploration of small molecule Legumain inhibitors with the aim of developing a potent and selective in vitro tool compound. Work continued in this Letter explores the use of alternative P2-P3 linker units and the P3 group SAR which led to the identification of 10t, a potent, selective and cellularly active Legumain inhibitor. We also demonstrate that 10t has activity in both cancer cell viability and colony formation assays.
Collapse
Affiliation(s)
- Kerry A Ness
- CCRCB Drug Discovery Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd., Belfast BT9 7BL, UK
| | - Sharon L Eddie
- CCRCB Drug Discovery Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd., Belfast BT9 7BL, UK
| | - Catherine A Higgins
- CCRCB Drug Discovery Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd., Belfast BT9 7BL, UK
| | - Amy Templeman
- CCRCB Drug Discovery Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd., Belfast BT9 7BL, UK
| | - Zenobia D'Costa
- CCRCB Drug Discovery Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd., Belfast BT9 7BL, UK
| | - Kishore K D Gaddale
- CCRCB Drug Discovery Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd., Belfast BT9 7BL, UK
| | - Samira Bouzzaoui
- CCRCB Drug Discovery Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd., Belfast BT9 7BL, UK
| | - Linda Jordan
- Almac Discovery, Almac House, 20 Seagoe Industrial Estate, Craigavon BT63 5QD, UK
| | - Dominic Janssen
- Almac Discovery, Almac House, 20 Seagoe Industrial Estate, Craigavon BT63 5QD, UK
| | - Timothy Harrison
- Almac Discovery, Almac House, 20 Seagoe Industrial Estate, Craigavon BT63 5QD, UK
| | - Frank Burkamp
- Almac Discovery, Almac House, 20 Seagoe Industrial Estate, Craigavon BT63 5QD, UK
| | - Andrew Young
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Rd., Belfast BT9 7BL, UK
| | - Roberta Burden
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Rd., Belfast BT9 7BL, UK
| | - Christopher J Scott
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Rd., Belfast BT9 7BL, UK
| | - Paul B Mullan
- CCRCB Drug Discovery Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd., Belfast BT9 7BL, UK
| | - Rich Williams
- CCRCB Drug Discovery Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Rd., Belfast BT9 7BL, UK.
| |
Collapse
|
28
|
Structure and function of legumain in health and disease. Biochimie 2015; 122:126-50. [PMID: 26403494 DOI: 10.1016/j.biochi.2015.09.022] [Citation(s) in RCA: 194] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 09/18/2015] [Indexed: 12/27/2022]
Abstract
The last years have seen a steady increase in our understanding of legumain biology that is driven from two largely uncoupled research arenas, the mammalian and the plant legumain field. Research on legumain, which is also referred to as asparaginyl endopeptidase (AEP) or vacuolar processing enzyme (VPE), is slivered, however. Here we summarise recent important findings and put them into a common perspective. Legumain is usually associated with its cysteine endopeptidase activity in lysosomes where it contributes to antigen processing for class II MHC presentation. However, newly recognized functions disperse previously assumed boundaries with respect to their cellular compartmentalisation and enzymatic activities. Legumain is also found extracellularly and even translocates to the cytosol and the nucleus, with seemingly incompatible pH and redox potential. These different milieus translate into changes of legumain's molecular properties, including its (auto-)activation, conformational stability and enzymatic functions. Contrasting its endopeptidase activity, legumain can develop a carboxypeptidase activity which remains stable at neutral pH. Moreover, legumain features a peptide ligase activity, with intriguing mechanistic peculiarities in plant and human isoforms. In pathological settings, such as cancer or Alzheimer's disease, the proper association of legumain activities with the corresponding cellular compartments is breached. Legumain's increasingly recognized physiological and pathological roles also indicate future research opportunities in this vibrant field.
Collapse
|
29
|
The ins and outs of MHC class II-mediated antigen processing and presentation. Nat Rev Immunol 2015; 15:203-16. [PMID: 25720354 DOI: 10.1038/nri3818] [Citation(s) in RCA: 662] [Impact Index Per Article: 73.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antigenic peptide-loaded MHC class II molecules (peptide-MHC class II) are constitutively expressed on the surface of professional antigen-presenting cells (APCs), including dendritic cells, B cells, macrophages and thymic epithelial cells, and are presented to antigen-specific CD4(+) T cells. The mechanisms of antigen uptake, the nature of the antigen processing compartments and the lifetime of cell surface peptide-MHC class II complexes can vary depending on the type of APC. It is likely that these differences are important for the function of each distinct APC subset in the generation of effective adaptive immune responses. In this Review, we describe our current knowledge of the mechanisms of uptake and processing of antigens, the intracellular formation of peptide-MHC class II complexes, the intracellular trafficking of peptide-MHC class II complexes to the APC plasma membrane and their ultimate degradation.
Collapse
|
30
|
Dall E, Fegg JC, Briza P, Brandstetter H. Struktur und Mechanismus einer Aspartimid-abhängigen Peptidligase in humanem Legumain. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201409135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
31
|
Dall E, Fegg JC, Briza P, Brandstetter H. Structure and mechanism of an aspartimide-dependent peptide ligase in human legumain. Angew Chem Int Ed Engl 2015; 54:2917-21. [PMID: 25630877 PMCID: PMC4506564 DOI: 10.1002/anie.201409135] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 11/04/2014] [Indexed: 02/01/2023]
Abstract
Peptide ligases expand the repertoire of genetically encoded protein architectures by synthesizing new peptide bonds, energetically driven by ATP or NTPs. Here, we report the discovery of a genuine ligase activity in human legumain (AEP) which has important roles in immunity and tumor progression that were believed to be due to its established cysteine protease activity. Defying dogma, the ligase reaction is independent of the catalytic cysteine but exploits an endogenous energy reservoir that results from the conversion of a conserved aspartate to a metastable aspartimide. Legumain's dual protease-ligase activities are pH- and thus localization controlled, dominating at acidic and neutral pH, respectively. Their relevance includes reversible on-off switching of cystatin inhibitors and enzyme (in)activation, and may affect the generation of three-dimensional MHC epitopes. The aspartate-aspartimide (succinimide) pair represents a new paradigm of coupling endergonic reactions in ATP-scarce environments.
Collapse
Affiliation(s)
- Elfriede Dall
- Department of Molecular Biology, University of Salzburg, 5020 Salzburg (Austria)
| | | | | | | |
Collapse
|
32
|
Hasegawa S, Yamasaki M, Fukui T. Degradation of acetoacetyl-CoA synthetase, a ketone body-utilizing enzyme, by legumain in the mouse kidney. Biochem Biophys Res Commun 2014; 453:631-5. [PMID: 25301556 DOI: 10.1016/j.bbrc.2014.09.130] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 09/30/2014] [Indexed: 01/12/2023]
Abstract
Acetoacetyl-CoA synthetase (AACS) is a ketone body-utilizing enzyme, which is responsible for the synthesis of cholesterol and fatty acids from ketone bodies in lipogenic tissues, such as the liver and adipocytes. To explore the possibility of AACS regulation at the protein-processing level, we investigated the proteolytic degradation of AACS. Western blot analysis showed that the 75.1kDa AACS was cleaved to form a protein of approximately 55kDa in the kidney, which has considerable high activity of legumain, a lysosomal asparaginyl endopeptidase. Co-expression of AACS and legumain in HEK 293 cells generated the 55kDa product from AACS. Moreover, incubation of recombinant AACS with recombinant legumain resulted in the degradation of AACS. Knockdown of legumain with short-hairpin RNA against legumain using the hydrodynamics method led to a decrease in the 55kDa band of AACS in mouse kidney. These results suggest that legumain is involved in the processing of AACS through the lysosomal degradation pathway in the kidney.
Collapse
Affiliation(s)
- Shinya Hasegawa
- Department of Health Chemistry, Hoshi University, Ebara, Shinagawa, Tokyo 142-8501, Japan.
| | - Masahiro Yamasaki
- Department of Health Chemistry, Hoshi University, Ebara, Shinagawa, Tokyo 142-8501, Japan
| | - Tetsuya Fukui
- Department of Health Chemistry, Hoshi University, Ebara, Shinagawa, Tokyo 142-8501, Japan
| |
Collapse
|
33
|
van Kasteren SI, Overkleeft HS. Endo-lysosomal proteases in antigen presentation. Curr Opin Chem Biol 2014; 23:8-15. [PMID: 25213682 DOI: 10.1016/j.cbpa.2014.08.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 02/04/2023]
Abstract
Endo-lysosomal proteases have long been attractive, yet elusive, targets for medicinal chemistry. They have found to play key roles in health and disease; with protease under- and over-activity having been implicated in cancer, osteoporosis and Alzheimer's disease. Here we will discuss their role in the adaptive immune response. The crucial roles of these enzymes multiple processes in antigen presentation will be discussed: from activating MHC-II receptors, to the production of epitopes from antigens and the activation of Toll-like receptors. The early efforts at pharmacological interventions in these pathways will also be discussed.
Collapse
Affiliation(s)
- Sander I van Kasteren
- Division of Chemical Biology, Leiden Institute of Chemistry, Leiden University, Gorlaeus Laboratory, The Netherlands.
| | - Herman S Overkleeft
- Division of Chemical Biology, Leiden Institute of Chemistry, Leiden University, Gorlaeus Laboratory, The Netherlands
| |
Collapse
|
34
|
Higgins C, Bouazzaoui S, Gaddale K, D’Costa Z, Templeman A, O’Rourke M, Young A, Scott C, Harrison T, Mullan P, Williams R. P3 SAR exploration of biphenyl carbamate based Legumain inhibitors. Bioorg Med Chem Lett 2014; 24:2521-4. [DOI: 10.1016/j.bmcl.2014.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 03/31/2014] [Accepted: 04/01/2014] [Indexed: 11/16/2022]
|
35
|
Clement CC, Santambrogio L. The lymph self-antigen repertoire. Front Immunol 2013; 4:424. [PMID: 24379811 PMCID: PMC3864156 DOI: 10.3389/fimmu.2013.00424] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 11/20/2013] [Indexed: 01/26/2023] Open
Abstract
The lymphatic fluid originates from the interstitial fluid which bathes every parenchymal organ and reflects the “omic” composition of the tissue from which it originates in its physiological or pathological signature. Several recent proteomic analyses have mapped the proteome-degradome and peptidome of this immunologically relevant fluid pointing to the lymph as an important source of tissue-derived self-antigens. A vast array of lymph-circulating peptides have been mapped deriving from a variety of processing pathways including caspases, cathepsins, MMPs, ADAMs, kallikreins, calpains, and granzymes, among others. These self peptides can be directly loaded on circulatory dendritic cells and expand the self-antigenic repertoire available for central and peripheral tolerance.
Collapse
Affiliation(s)
- Cristina C Clement
- Department of Pathology, Albert Einstein College of Medicine , New York, NY , USA ; Department of Microbiology and Immunology, Albert Einstein College of Medicine , New York, NY , USA
| | - Laura Santambrogio
- Department of Pathology, Albert Einstein College of Medicine , New York, NY , USA ; Department of Microbiology and Immunology, Albert Einstein College of Medicine , New York, NY , USA
| |
Collapse
|
36
|
van Kasteren SI, Overkleeft H, Ovaa H, Neefjes J. Chemical biology of antigen presentation by MHC molecules. Curr Opin Immunol 2013; 26:21-31. [PMID: 24556397 DOI: 10.1016/j.coi.2013.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 10/03/2013] [Indexed: 11/25/2022]
Abstract
MHC class I and MHC class II molecules present peptides to the immune system to drive proper T cell responses. Pharmacological modulation of T-cell responses can offer treatment options for a range of immune-related diseases. Pharmacological downregulation of MHC molecules may find application in treatment of auto-immunity and transplantation rejection while pharmacological activation of antigen presentation would support immune responses to infection and cancer. Since the cell biology of MHC class I and MHC class II antigen presentation is understood in great detail, many potential targets for manipulation have been defined over the years. Here, we discuss how antigen presentation by MHC molecules can be modulated by pharmacological agents and how chemistry can further support the study of antigen presentation in general. The chemical biology of antigen presentation by MHC molecules shows surprising options for immune modulation and the development of future therapies.
Collapse
Affiliation(s)
- Sander I van Kasteren
- The Netherlands Cancer Institute, Division of Cell Biology, Plesmanlaan 121, Amsterdam, Netherlands; Leiden Institute of Chemistry (LIC), Division of Bio-Organic Chemistry, Einsteinweg 55, Leiden, Netherlands
| | - Hermen Overkleeft
- The Netherlands Cancer Institute, Division of Cell Biology, Plesmanlaan 121, Amsterdam, Netherlands; Leiden Institute of Chemistry (LIC), Division of Bio-Organic Chemistry, Einsteinweg 55, Leiden, Netherlands
| | - Huib Ovaa
- The Netherlands Cancer Institute, Division of Cell Biology, Plesmanlaan 121, Amsterdam, Netherlands; Leiden Institute of Chemistry (LIC), Division of Bio-Organic Chemistry, Einsteinweg 55, Leiden, Netherlands
| | - Jacques Neefjes
- The Netherlands Cancer Institute, Division of Cell Biology, Plesmanlaan 121, Amsterdam, Netherlands; Leiden Institute of Chemistry (LIC), Division of Bio-Organic Chemistry, Einsteinweg 55, Leiden, Netherlands.
| |
Collapse
|
37
|
Manoury B. Proteases: essential actors in processing antigens and intracellular toll-like receptors. Front Immunol 2013; 4:299. [PMID: 24065969 PMCID: PMC3781364 DOI: 10.3389/fimmu.2013.00299] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 09/09/2013] [Indexed: 01/28/2023] Open
Abstract
MHC class II molecules expressed by professional antigen presenting cells (pAPCs) such as macrophages, B cells, and dendritic cells (DCs) play a fundamental role in presenting peptides to CD4+ T cells. However, to elicit CD4+-T cells immunity, pAPCs need an additional signal, which can be delivered by toll-like receptors (TLRs) molecules. TLRs recognize microbial patterns and are critical in initiating immune responses. Proteases, which provide peptide ligands for the MHC class II antigenic presentation pathway, were recently shown to cleave and activate intracellular TLRs in endosomal compartments. Here, I give an overview on the individual roles of the most well studied proteases in both antigen and TLRs processing.
Collapse
Affiliation(s)
- Bénédicte Manoury
- Institut National de la Santé et de le Recherche Médicale, Unité 1013, Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine , Paris , France
| |
Collapse
|
38
|
Bourgeois-Daigneault MC, Thibodeau J. Identification of a novel motif that affects the conformation and activity of the MARCH1 E3 ubiquitin ligase. J Cell Sci 2012; 126:989-98. [PMID: 23264739 DOI: 10.1242/jcs.117804] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
MARCH1, a member of the membrane-associated RING-CH family of E3 ubiquitin ligases, regulates antigen presentation by downregulating the cell surface expression of Major Histocompatibility Complex class II and CD86 molecules. MARCH1 is a transmembrane protein that exposes both its N- and C-terminus to the cytoplasm. We have conducted a structure-function analysis of its two cytoplasmic tails to gain insights into the trafficking of MARCH1 in the endocytic pathway. Fusion of the N-terminal portion of MARCH1 to a type II transmembrane reporter molecule revealed that this cytoplasmic tail contains endosomal sorting motifs. The C-terminal domain also appears to contain intracellular sorting signals because it reduced surface expression of a type I transmembrane reporter molecule. Mutation of the two putative C-terminal tyrosine-based sorting signals did not affect the activity of human MARCH1; however, it did reduce its incorporation into exosomes. Moreover, site-directed mutagenesis pointed to a functional C-terminal 221VQNC224 sequence that affects the spatial organization of the two cytoplasmic regions. This motif is also found in other RING-type E3 ubiquitin ligases, such as parkin. Altogether, these findings highlight the complex regulation of MARCH1 trafficking in the endocytic pathway as well as the intricate interactions between its cytoplasmic tails.
Collapse
|
39
|
Edgington LE, Verdoes M, Ortega A, Withana NP, Lee J, Syed S, Bachmann MH, Blum G, Bogyo M. Functional imaging of legumain in cancer using a new quenched activity-based probe. J Am Chem Soc 2012; 135:174-82. [PMID: 23215039 DOI: 10.1021/ja307083b] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Legumain is a lysosomal cysteine protease whose biological function remains poorly defined. Legumain activity is up-regulated in most human cancers and inflammatory diseases most likely as the result of high expression in populations of activated macrophages. Within the tumor microenvironment, legumain activity is thought to promote tumorigenesis. To obtain a greater understanding of the role of legumain activity during cancer progression and inflammation, we developed an activity-based probe that becomes fluorescent only upon binding active legumain. This probe is highly selective for legumain, even in the context of whole cells and tissues, and is also a more effective label of legumain than previously reported probes. Here we present the synthesis and application of our probe to the analysis of legumain activity in primary macrophages and in two mouse models of cancer. We find that legumain activity is highly correlated with macrophage activation and furthermore that it is an ideal marker for primary tumor inflammation and early stage metastatic lesions.
Collapse
Affiliation(s)
- Laura E Edgington
- Cancer Biology Program, Department of Pathology, Stanford School of Medicine, 300 Pasteur Drive, Stanford, California 94305-5324, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sandberg A, Lindell G, Källström BN, Branca RM, Danielsson KG, Dahlberg M, Larson B, Forshed J, Lehtiö J. Tumor proteomics by multivariate analysis on individual pathway data for characterization of vulvar cancer phenotypes. Mol Cell Proteomics 2012; 11:M112.016998. [PMID: 22499770 DOI: 10.1074/mcp.m112.016998] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Vulvar squamous cell carcinoma (VSCC) is the fourth most common gynecological cancer. Based on etiology VSCC is divided into two subtypes; one related to high-risk human papilloma virus (HPV) and one HPV negative. The two subtypes are proposed to develop via separate intracellular signaling pathways. We investigated a suggested link between HPV infection and relapse risk in VSCC through in-depth protein profiling of 14 VSCC tumor specimens. The tumor proteomes were analyzed by liquid-chromatography tandem mass spectrometry. Relative protein quantification was performed by 8-plex isobaric tags for relative and absolute quantification. Labeled peptides were fractionated by high-resolution isoelectric focusing prior to liquid-chromatography tandem mass spectrometry to reduce sample complexity. In total, 1579 proteins were regarded as accurately quantified and analyzed further. For classification of clinical groups, data analysis was performed by comparing protein level differences between tumors defined by HPV and/or relapse status. Further, we performed a biological analysis on individual tumor proteomes by matching data to known biological pathways. We here present a novel analysis approach that combines pathway alteration data on individual tumor level with multivariate statistics for HPV and relapse status comparisons. Four proteins (signal transducer and activator of transcription-1, myxovirus resistance protein 1, proteasome subunit alpha type-5 and legumain) identified as main classifiers of relapse status were validated by immunohistochemistry (IHC). Two of the proteins are interferon-regulated and on mRNA level known to be repressed by HPV. By both liquid-chromatography tandem mass spectrometry and immunohistochemistry data we could single out a subgroup of HPV negative/relapse-associated tumors. The pathway level data analysis confirmed three of the proteins, and further identified the ubiquitin-proteasome pathway as altered in the high risk subgroup. We show that pathway fingerprinting with resolution on individual tumor level adds biological information that strengthens a generalized protein analysis.
Collapse
Affiliation(s)
- Annsofi Sandberg
- Clinical Proteomics Mass Spectrometry, Department of Oncology-Pathology, Science for Life Laboratory and Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Majera D, Kristan KČ, Neefjes J, Turk D, Mihelič M. Expression, purification and assembly of soluble multimeric MHC class II-invariant chain complexes. FEBS Lett 2012; 586:1318-24. [PMID: 22465664 DOI: 10.1016/j.febslet.2012.03.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 03/14/2012] [Indexed: 11/15/2022]
Abstract
Major histocompatibility class (MHC) II molecules are essential for running adaptive immune response. They are produced in the ER and targeted to late endosomes with the help of invariant chain (Ii) trimers. Ii trimerization may be induced by the Ii TM domain. To enable mechanistic and structural studies of MHC class II-Ii assembly, soluble forms of the complexes were expressed. We show that Ii trimerizes in the absence of the transmembrane part, prior to binding of α/β chains. The biochemical analysis supports the suggestion that the MHC class II-Ii complexes are not necessarily trimers of trimers, but that the Ii trimer can also be occupied by one or two MHC class II complexes.
Collapse
Affiliation(s)
- Dušana Majera
- Department of Biochemistry, Molecular and Structural Biology and Centre for Protein and Structure Production, Jozef Stefan Institute, Ljubljana, Slovenia
| | | | | | | | | |
Collapse
|
42
|
Lee J, Bogyo M. Synthesis and evaluation of aza-peptidyl inhibitors of the lysosomal asparaginyl endopeptidase, legumain. Bioorg Med Chem Lett 2011; 22:1340-3. [PMID: 22243962 DOI: 10.1016/j.bmcl.2011.12.079] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 12/13/2011] [Accepted: 12/14/2011] [Indexed: 11/16/2022]
Abstract
Legumain or asparaginly endopeptidase (AEP) is a lysosomal cysteine protease with a high level of specificity for cleavage of protein substrates after an asparagine residue. It is also capable of cleaving after aspartic acids sites when in the acidic environment of the lysosome. Legumain expression and activity is linked to a number of pathological conditions including cancer, atherosclerosis and inflammation, yet its biological role in these pathologies is not well-understood. Highly potent and selective inhibitors of legumain would not only be valuable for studying the functional roles of legumain in these conditions, but may have therapeutic potential as well. We describe here the design, synthesis and in vitro evaluation of selective legumain inhibitors based on the aza-asparaginyl scaffold. We synthesized a library of aza-peptidyl inhibitors with various non-natural amino acids and different electrophilic warheads, and characterized the kinetic properties of inactivation of legumain. We also synthesized fluorescently labeled inhibitors to investigate cell permeability and selectivity of the compounds. The inhibitors have second order rate constants of up to 5 × 10(4)M(-1)s(-1) and IC(50) values as low as 4 nM against recombinant mouse legumain. In addition, the inhibitors are highly selective toward legumain and have little or no cross-reactivity with cathepsins. Overall, we have identified several valuable new inhibitors of legumain that can be used to study legumain function in multiple disease models.
Collapse
Affiliation(s)
- Jiyoun Lee
- Department of Pathology and Microbiology and Immunology, Stanford University School of Medicine, 300 Pasteur Dr. Stanford, CA 94305-5324, USA.
| | | |
Collapse
|
43
|
Nucleoplasmic calcium regulates cell proliferation through legumain. J Hepatol 2011; 55:626-635. [PMID: 21237226 PMCID: PMC3158841 DOI: 10.1016/j.jhep.2010.12.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 12/09/2010] [Accepted: 12/11/2010] [Indexed: 01/19/2023]
Abstract
BACKGROUND & AIMS Nucleoplasmic Ca(2+) regulates cell growth in the liver, but the proteins through which this occurs are unknown. METHODS We used Rapid Subtraction Hybridization (RaSH) to subtract genes in SKHep1 liver cells expressing the Ca(2+) buffer protein parvalbumin (PV) targeted to the nucleus, from genes in cells expressing a mutated form of nuclear-targeted PV which has one of two Ca(2+)-binding sites inactivated. The subtraction permitted the selection of genes whose expression was affected by a small alteration in nuclear Ca(2+) concentration. RESULTS The asparaginyl endopeptidase legumain (LGMN) was identified in this screening. When Ca(2+) was buffered in the nucleus of SKHep1 cells, LGMN mRNA was decreased by 97%, in part by a transcriptional mechanism, and decreased expression at the protein level was observed by immunoblot and immunofluorescence. Treatment with hepatocyte growth factor increased LGMN expression. Knockdown of LGMN by siRNA decreased proliferation of SKHep1 cells by ∼50% as measured both by BrdU uptake and mitotic index, although an inhibitor of LGMN activity did not affect BrdU incorporation. A significant reduction in the fraction of cells in G2/M phase was seen as well. This was associated with increases in the expression of cyclins A and E. Furthermore, LGMN expression was increased in hepatocellular carcinoma cells relative to normal hepatocytes in the same specimens. CONCLUSIONS These findings suggest a new role for LGMN and provide evidence that nuclear Ca(2+) signals regulate cell proliferation in part through the modulation of LGMN expression. Increased expression of LGMN may be involved in liver carcinogenesis.
Collapse
|
44
|
Müller S, Dennemärker J, Reinheckel T. Specific functions of lysosomal proteases in endocytic and autophagic pathways. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2011; 1824:34-43. [PMID: 21767668 PMCID: PMC7105187 DOI: 10.1016/j.bbapap.2011.07.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 07/04/2011] [Accepted: 07/05/2011] [Indexed: 12/17/2022]
Abstract
Endolysosomal vesicles form a highly dynamic multifunctional cellular compartment that contains multiple highly potent proteolytic enzymes. Originally these proteases have been assigned to cooperate solely in executing the unselective ‘bulk proteolysis’ within the acidic milieu of the lysosome. Although to some degree this notion still holds true, evidence is accumulating for specific and regulatory functions of individual ‘acidic’ proteases in many cellular processes linked to the endosomal/lysosomal compartment. Here we summarize and discuss the functions of individual endolysosomal proteases in such diverse processes as the termination of growth factor signaling, lipoprotein particle degradation, infection, antigen presentation, and autophagy. This article is part of a Special Issue entitled: Proteolysis 50 years after the discovery of lysosome.
Collapse
Affiliation(s)
- Sabrina Müller
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Freiburg, D-79104, Germany
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Julia Dennemärker
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Freiburg, D-79104, Germany
- Dept. of Visceral Surgery, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Freiburg, D-79104, Germany
- BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- Corresponding author at: Institut für Molekulare Medizin und Zellforschung, Albert-Ludwigs-Universität-Freiburg, Stefan Meier Str. 17, D-79104 Freiburg, Germany. Tel.: + 49 761 203 9606; fax: + 49 761 203 9634.
| |
Collapse
|
45
|
Bajjuri KM, Liu Y, Liu C, Sinha SC. The legumain protease-activated auristatin prodrugs suppress tumor growth and metastasis without toxicity. ChemMedChem 2011; 6:54-9. [PMID: 21154805 DOI: 10.1002/cmdc.201000478] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Krishna Mohan Bajjuri
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
46
|
Reich M, Zou F, Sieńczyk M, Oleksyszyn J, Boehm BO, Burster T. Invariant chain processing is independent of cathepsin variation between primary human B cells/dendritic cells and B-lymphoblastoid cells. Cell Immunol 2011; 269:96-103. [PMID: 21543057 DOI: 10.1016/j.cellimm.2011.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 02/16/2011] [Accepted: 03/13/2011] [Indexed: 01/01/2023]
Abstract
As part of the endocytic antigen processing pathway, proteolytic cleavage of the invariant chain (Ii) is important for the generation of class II-associated invariant chain peptide (CLIP). CLIP remains associated with the major histocompatibility complex (MHC) class II molecule to prevent premature loading of antigenic peptides. Cysteine proteases, such as Cathepsin S (CatS), CatL, or CatV, play a pivotal role in the final stage of Ii degradation depending on the cell type studied. Less is known regarding the early stages of Ii processing. We therefore explored whether the serine protease CatG is involved in the initial step of Ii degradation in primary antigen presenting cells (APC), since the cathepsin distribution differs between primary APC and cell lines. While primary human B cells and dendritic cells (DC) do harbor CatG, this protease is absent in B-lymphoblastoid cells (BLC) or monocyte-derived DC generated in vitro. In addition, other proteases, such as CatC, CatL, and the asparagine endoprotease (AEP), are active in BLC and monocyte-derived DC. Here we demonstrate that CatG progressively degraded Ii in vitro resulting in several intermediates. However, pharmacological inhibition of CatG in primary B cells and DC did not alter Ii processing, indicating that CatG is dispensable in Ii degradation. Interestingly, stalling of cysteine proteases by inhibition in BLC vs. primary B cells and DC did not result in any differences in the generation of distinct Ii intermediates between the cells tested, suggesting that Ii processing is independent of the cathepsin variation within professional human APC.
Collapse
Affiliation(s)
- Michael Reich
- Division of Endocrinology and Diabetes, Center for Internal Medicine, University Medical Center Ulm, Ulm, Germany
| | | | | | | | | | | |
Collapse
|
47
|
Miller G, Matthews SP, Reinheckel T, Fleming S, Watts C. Asparagine endopeptidase is required for normal kidney physiology and homeostasis. FASEB J 2011; 25:1606-17. [DOI: 10.1096/fj.10-172312] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Gail Miller
- Division of Cell Biology and ImmunologyCollege of Life SciencesUniversity of DundeeDundeeUK
| | - Stephen P. Matthews
- Division of Cell Biology and ImmunologyCollege of Life SciencesUniversity of DundeeDundeeUK
| | - Thomas Reinheckel
- Institut für Molekulare Medizin und ZellforschungAlbert-Ludwigs-Universität FreiburgFreiburgGermany
| | - Stewart Fleming
- Department of Molecular PathologyNinewells HospitalUniversity of DundeeDundeeUK
| | - Colin Watts
- Division of Cell Biology and ImmunologyCollege of Life SciencesUniversity of DundeeDundeeUK
| |
Collapse
|
48
|
Borghese F, Clanchy FIL. CD74: an emerging opportunity as a therapeutic target in cancer and autoimmune disease. Expert Opin Ther Targets 2011; 15:237-51. [PMID: 21208136 DOI: 10.1517/14728222.2011.550879] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION CD74, also known as the invariant chain, participates in several key processes of the immune system, including antigen presentation, B-cell differentiation and inflammatory signaling. Despite being described more than 3 decades ago, new functions and novel interactions for this evolutionarily conserved molecule are still being unraveled. As a participant in several immunological processes and an indicator of disease in some conditions, it has potential as a therapeutic target. AREAS COVERED The relationship between the structure of CD74 variants and their physiological functions is detailed in this review. The function of CD74 in several cell lineages is examined with a focus on the interactions with cathepsins and, in an inflammatory milieu, the pro-inflammatory cytokine macrophage migratory inhibitory factor. The role of CD74 signaling in inflammatory and carcinogenic processes is outlined as is the use of CD74 as a therapeutic target (in cancer) and tool (as a vaccine). EXPERT OPINION CD74 has several roles within the cell and throughout the immune system. Most prominent amongst these are the complex relationships with MIF and cathepsins. Modulation of CD74 function shows promise for the effective amelioration of disease.
Collapse
Affiliation(s)
- Federica Borghese
- Sapienza University of Rome, Department of Clinical Medicine, Clinical Immunology Unit, Umberto I Policlinico di Roma, 155 Viale del Policlinico, Rome, IT 00161
| | | |
Collapse
|
49
|
Matthews SP, Werber I, Deussing J, Peters C, Reinheckel T, Watts C. Distinct protease requirements for antigen presentation in vitro and in vivo. THE JOURNAL OF IMMUNOLOGY 2010; 184:2423-31. [PMID: 20164435 DOI: 10.4049/jimmunol.0901486] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Asparagine endopeptidase (AEP) or legumain is a potentially important Ag-processing enzyme that introduces limited cleavages that trigger unfolding and class II MHC binding of different Ag substrates. AEP is necessary and sufficient for optimal processing and presentation of the tetanus toxin C fragment (TTCF) Ag in vitro, but its importance has not been tested in vivo. Surprisingly, virtually normal T cell and Ab responses to TTCF were mounted in AEP-deficient mice when examined 10 d after immunization. This was the case when TTCF was emulsified with CFA, adsorbed onto alum, or expressed within live Salmonella typhimurium. In addition, the dominant Ab and T cell determinants recognized in TTCF were essentially unchanged in AEP-deficient mice. These data are explained, at least in part, by the much lower levels of AEP expressed in primary murine APCs compared with immortalized B cell lines. Even so, the initial in vivo kinetics of TTCF presentation were slower in AEP-deficient mice and, as expected, boosting AEP levels in primary APCs enhanced and accelerated TTCF processing and presentation in vitro. Thus, AEP remains the protease of choice for TTCF processing; however, in its absence, other enzymes can substitute to enable slower, but equally robust, adaptive immune responses. Moreover, clear relationships between Ags and processing proteases identified from short-term in vitro processing and presentation studies do not necessarily predict an absolute in vivo dependency on those processing enzymes, not least because they may be expressed at strikingly different levels in vitro versus in vivo.
Collapse
Affiliation(s)
- Stephen P Matthews
- Division of Cell Biology and Immunology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | | | | | | | | | | |
Collapse
|
50
|
Berkova Z, Tao RH, Samaniego F. Milatuzumab - a promising new immunotherapeutic agent. Expert Opin Investig Drugs 2010; 19:141-9. [PMID: 19968579 DOI: 10.1517/13543780903463854] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Milatuzumab is a new immunotherapeutic agent targeting CD74, a membrane protein preferentially expressed in hematopoietic cancers and some solid tumors. Broad expression and fast internalization makes CD74 an ideal target for cancer therapy. We reviewed published articles about CD74 and milatuzumab. We present a comprehensive review of CD74 functions and provide explanation of milatuzumab antitumor effects. This review describes CD74 protein biology with the emphasis on the role of CD74 in tumor survival and its new role in regulation of the Fas death receptor. The development of CD74 targeting therapies to induce tumor regression and cancer cell apoptosis is described and results of clinical trials are discussed. Milatuzumab shows selective binding and rapid internalization into CD74-positive cancer cells. Milatuzumab with and without conjugated toxins synergizes with other chemotherapeutic agents and elicits significant antitumor effects in mice. In a Phase I trial, milatuzumab showed no severe adverse effects in patients with relapsed/refractory multiple myeloma and it stabilized the disease in some patients for up to 12 weeks. Ongoing trials testing different treatment schedules of milatuzumab in chronic lymphocytic leukemia, non-Hodgkin's lymphoma and multiple myeloma indicate that milatuzumab shows no severe adverse effects in humans.
Collapse
Affiliation(s)
- Zuzana Berkova
- The University of Texas, MD Anderson Cancer Center, Department of Lymphoma/Myeloma, Houston, Texas 77030, USA
| | | | | |
Collapse
|