1
|
Liao ZC, Yang MC, Xie DR, Abbas F, Zeng RF, Huang XM, Wang HC. Targeting a cysteine proteinase inhibitor and a defensin-like protein in Litchi chinensis seed development leveraging endosperm single-nucleus transcriptome. Int J Biol Macromol 2025; 296:139708. [PMID: 39798746 DOI: 10.1016/j.ijbiomac.2025.139708] [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: 11/02/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Seed development is one of the most important agricultural traits, determining both the crop yield and quality of fleshy fruits. A typically abortive litchi cultivar, Guiwei, exhibits heterogeneity in seed size across production areas, years, and individual trees. Previous studies have shown that 'Guiwei' seed development failure is associated with endosperm arrest and chilling conditions. Herein, we identified a cysteine proteinase inhibitor (LcCPI5) and defensin-like protein (LcDEFL) as key genes in determining 'Guiwei' seed development through combined analysis of 'Guiwei' and 'Huaizhi' endosperm single-nucleus RNA-sequence and transcriptome data of 'Guiwei' seed subjected to different temperature treatments. LcCPI5 was exclusively expressed in the endosperm sample of 'Guiwei' at 25 days post-anthesis, and its expression decreased in response to chilling. The silencing of LcCPI5 led to significantly larger seeds, whereas employing the cysteine proteinase inhibitor E-64 resulted in smaller seeds in the cultivar 'Guiwei'. Unlike LcCPI5, LcDEFL promotes litchi seed development. The large seed cultivar 'Huaizhi' had substantially higher expression of LcDEFL than the partly abortive cultivar 'Guiwei'. LcDEFL silencing led to a notable reduction in the size of litchi seeds. These findings point to the post-translational modulation of cysteine proteinase and the critical role of cysteine-rich proteins in litchi seed development.
Collapse
Affiliation(s)
- Zhi-Chan Liao
- Key Laboratory of Biology and Genetic Improvement of Horticultural Crops-South China/Guangdong Litchi Engineering Research Center, College of Horticulture, South China Agricultural University, Guangzhou, China.
| | - Ming-Chao Yang
- Institute of Tropical Fruit Trees, Hainan Academy of Agricultural Sciences/Key Laboratory of Genetic Resources and Utilization of Tropical Fruits and Vegetables, Haikou 571100, Hainan, China
| | - Dan-Rong Xie
- Key Laboratory of Biology and Genetic Improvement of Horticultural Crops-South China/Guangdong Litchi Engineering Research Center, College of Horticulture, South China Agricultural University, Guangzhou, China
| | - Farhat Abbas
- Key Laboratory of Biology and Genetic Improvement of Horticultural Crops-South China/Guangdong Litchi Engineering Research Center, College of Horticulture, South China Agricultural University, Guangzhou, China; Institute of Tropical Fruit Trees, Hainan Academy of Agricultural Sciences/Key Laboratory of Genetic Resources and Utilization of Tropical Fruits and Vegetables, Haikou 571100, Hainan, China
| | - Ren-Fang Zeng
- Key Laboratory of Biology and Genetic Improvement of Horticultural Crops-South China/Guangdong Litchi Engineering Research Center, College of Horticulture, South China Agricultural University, Guangzhou, China.
| | - Xu-Ming Huang
- Key Laboratory of Biology and Genetic Improvement of Horticultural Crops-South China/Guangdong Litchi Engineering Research Center, College of Horticulture, South China Agricultural University, Guangzhou, China.
| | - Hui-Cong Wang
- Key Laboratory of Biology and Genetic Improvement of Horticultural Crops-South China/Guangdong Litchi Engineering Research Center, College of Horticulture, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
2
|
Forbord KM, Lunde NN, Bosnjak-Olsen T, Johansen HT, Solberg R, Jafari A. Legumain is a paracrine regulator of osteoblast differentiation and mediates the inhibitory effect of TGF-β1 on osteoblast maturation. Front Endocrinol (Lausanne) 2024; 15:1445049. [PMID: 39363898 PMCID: PMC11446771 DOI: 10.3389/fendo.2024.1445049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024] Open
Abstract
Transforming growth factor-beta 1 (TGF-β1) is a critical regulator of skeletal homeostasis and has diverse effects on osteoblastogenesis. To date, the mechanisms behind the intriguing inhibitory effect of TGF-β1 on osteoblast maturation are not fully understood. Here, we demonstrate a novel mechanism by which TGF-β1 modulates osteoblast maturation through the lysosomal protease legumain. We observed that addition of TGF-β1 to osteogenic cultures of human bone marrow derived mesenchymal stromal (stem) cells enhanced legumain activity and secretion, in-spite of decreased legumain mRNA expression, suggesting post-transcriptional regulation. We further showed that osteogenic cells internalize and activate prolegumain, associated with inhibited osteoblast maturation, revealing legumain as a paracrine regulator of osteoblast maturation. Interestingly, TGF-β1 treatment exacerbated legumain internalization and activity, and showed an additive effect on legumain-induced inhibition of osteoblast maturation. Importantly, pharmacological inhibition of legumain abolished the inhibitory effect of TGF-β1 on osteoblast maturation. Our findings reveal that TGF-β1 inhibits osteoblast maturation by stimulating secretion and activity of endogenous legumain, as well as enhancing internalization and activation of extracellular prolegumain. Therefore, our study provides a deeper understanding of the complex regulation of osteoblastogenesis and unveils a novel TGF-β1-legumain axis in regulation of osteoblast maturation, offering novel insights for possible therapeutic interventions related to bone diseases associated with aberrant TGF-β1 signaling.
Collapse
Affiliation(s)
- Karl Martin Forbord
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Ngoc Nguyen Lunde
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Tatjana Bosnjak-Olsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Harald Thidemann Johansen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Rigmor Solberg
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Wang Y, Peng Y, Zi G, Chen J, Peng B. Co-delivery of Cas9 mRNA and guide RNAs for editing of LGMN gene represses breast cancer cell metastasis. Sci Rep 2024; 14:8095. [PMID: 38582932 PMCID: PMC10998893 DOI: 10.1038/s41598-024-58765-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 04/03/2024] [Indexed: 04/08/2024] Open
Abstract
Legumain (or asparagine endopeptidase/AEP) is a lysosomal cysteine endopeptidase associated with increased invasive and migratory behavior in a variety of cancers. In this study, co-delivery of Cas9 mRNA and guide RNA (gRNA) by lipid nanoparticles (LNP) for editing of LGMN gene was performed. For in-vitro transcription (IVT) of gRNA, two templates were designed: linearized pUC57-T7-gRNA and T7-gRNA oligos, and the effectiveness of gRNA was verified in multiple ways. Cas9 plasmid was modified and optimized for IVT of Cas9 mRNA. The effects of LGMN gene editing on lysosomal/autophagic function and cancer cell metastasis were investigated. Co-delivery of Cas9 mRNA and gRNA resulted in impaired lysosomal/autophagic degradation, clone formation, migration, and invasion capacity of cancer cells in-vitro. Experimental lung metastasis experiment indicates co-delivery of Cas9 mRNA and gRNA by LNP reduced the migration and invasion capacity of cancer cells in-vivo. These results indicate that co-delivery of Cas9 mRNA and gRNA can enhance the efficiency of CRISPR/Cas9-mediated gene editing in-vitro and in-vivo, and suggest that Cas9 mRNA and gRNA gene editing of LGMN may be a potential treatment for breast tumor metastasis.
Collapse
Affiliation(s)
- Yue Wang
- College of Pharmacy, Dali University, 2 HongShen Road, Dali, 671003, Yunnan, China
| | - Yatu Peng
- JinCai High School, 2788 Yang Gao Middle Road, Pudong New District, Shanghai, 200135, China
| | - Guanghui Zi
- College of Pharmacy, Dali University, 2 HongShen Road, Dali, 671003, Yunnan, China
| | - Jin Chen
- College of Pharmacy, Dali University, 2 HongShen Road, Dali, 671003, Yunnan, China
| | - Baowei Peng
- College of Pharmacy, Dali University, 2 HongShen Road, Dali, 671003, Yunnan, China.
| |
Collapse
|
4
|
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
|
5
|
Euceda-Padilla EA, Mateo-Cruz MG, Ávila-González L, Flores-Pucheta CI, Ortega-López J, Talamás-Lara D, Velazquez-Valassi B, Jasso-Villazul L, Arroyo R. Trichomonas vaginalis Legumain-2, TvLEGU-2, Is an Immunogenic Cysteine Peptidase Expressed during Trichomonal Infection. Pathogens 2024; 13:119. [PMID: 38392857 PMCID: PMC10892250 DOI: 10.3390/pathogens13020119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Trichomonas vaginalis is the causative agent of trichomoniasis, the most prevalent nonviral, neglected sexually transmitted disease worldwide. T. vaginalis has one of the largest degradomes among unicellular parasites. Cysteine peptidases (CPs) are the most abundant peptidases, constituting 50% of the degradome. Some CPs are virulence factors recognized by antibodies in trichomoniasis patient sera, and a few are found in vaginal secretions that show fluctuations in glucose concentrations during infection. The CPs of clan CD in T. vaginalis include 10 genes encoding legumain-like peptidases of the C13 family. TvLEGU-2 is one of them and has been identified in multiple proteomes, including the immunoproteome obtained with Tv (+) patient sera. Thus, our goals were to assess the effect of glucose on TvLEGU-2 expression, localization, and in vitro secretion and determine whether TvLEGU-2 is expressed during trichomonal infection. We performed qRT-PCR assays using parasites grown under different glucose conditions. We also generated a specific anti-TvLEGU-2 antibody against a synthetic peptide of the most divergent region of this CP and used it in Western blot (WB) and immunolocalization assays. Additionally, we cloned and expressed the tvlegu-2 gene (TVAG_385340), purified the recombinant TvLEGU-2 protein, and used it as an antigen for immunogenicity assays to test human sera from patients with vaginitis. Our results show that glucose does not affect tvlegu-2 expression but does affect localization in different parasite organelles, such as the plasma membrane, Golgi complex, hydrogenosomes, lysosomes, and secretion vesicles. TvLEGU-2 is secreted in vitro, is present in vaginal secretions, and is immunogenic in sera from Tv (+) patients, suggesting its relevance during trichomonal infection.
Collapse
Affiliation(s)
- Esly Alejandra Euceda-Padilla
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, Alcaldía Gustavo A. Madero (GAM), Mexico City 07360, Mexico; (E.A.E.-P.); (M.G.M.-C.); (L.Á.-G.)
| | - Miriam Guadalupe Mateo-Cruz
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, Alcaldía Gustavo A. Madero (GAM), Mexico City 07360, Mexico; (E.A.E.-P.); (M.G.M.-C.); (L.Á.-G.)
| | - Leticia Ávila-González
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, Alcaldía Gustavo A. Madero (GAM), Mexico City 07360, Mexico; (E.A.E.-P.); (M.G.M.-C.); (L.Á.-G.)
| | - Claudia Ivonne Flores-Pucheta
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, Alcaldía Gustavo A. Madero (GAM), Mexico City 07360, Mexico; (C.I.F.-P.); (J.O.-L.)
| | - Jaime Ortega-López
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, Alcaldía Gustavo A. Madero (GAM), Mexico City 07360, Mexico; (C.I.F.-P.); (J.O.-L.)
| | - Daniel Talamás-Lara
- Unidad de Microscopía Electrónica, Laboratorios Nacionales De Servicios Experimentales (LaNSE), Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, Alcaldía Gustavo A. Madero (GAM), Mexico City 07360, Mexico;
| | - Beatriz Velazquez-Valassi
- Departamento de Vigilancia Epidemiológica, Hospital General de México “Eduardo Liceaga”, Mexico City 06720, Mexico;
| | - Lidia Jasso-Villazul
- Unidad de Medicina Preventiva, Hospital General de México “Eduardo Liceaga”, Mexico City 06720, Mexico;
| | - Rossana Arroyo
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, Alcaldía Gustavo A. Madero (GAM), Mexico City 07360, Mexico; (E.A.E.-P.); (M.G.M.-C.); (L.Á.-G.)
| |
Collapse
|
6
|
Santos NP, Soh WT, Demir F, Tenhaken R, Briza P, Huesgen PF, Brandstetter H, Dall E. Phytocystatin 6 is a context-dependent, tight-binding inhibitor of Arabidopsis thaliana legumain isoform β. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2023; 116:1681-1695. [PMID: 37688791 PMCID: PMC10952133 DOI: 10.1111/tpj.16458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/11/2023]
Abstract
Plant legumains are crucial for processing seed storage proteins and are critical regulators of plant programmed cell death. Although research on legumains boosted recently, little is known about their activity regulation. In our study, we used pull-down experiments to identify AtCYT6 as a natural inhibitor of legumain isoform β (AtLEGβ) in Arabidopsis thaliana. Biochemical analysis revealed that AtCYT6 inhibits both AtLEGβ and papain-like cysteine proteases through two separate cystatin domains. The N-terminal domain inhibits papain-like proteases, while the C-terminal domain inhibits AtLEGβ. Furthermore, we showed that AtCYT6 interacts with legumain in a substrate-like manner, facilitated by a conserved asparagine residue in its reactive center loop. Complex formation was additionally stabilized by charged exosite interactions, contributing to pH-dependent inhibition. Processing of AtCYT6 by AtLEGβ suggests a context-specific regulatory mechanism with implications for plant physiology, development, and programmed cell death. These findings enhance our understanding of AtLEGβ regulation and its broader physiological significance.
Collapse
Affiliation(s)
- Naiá P. Santos
- Department of Biosciences and Medical BiologyUniversity of Salzburg5020SalzburgAustria
| | - Wai Tuck Soh
- Department of Biosciences and Medical BiologyUniversity of Salzburg5020SalzburgAustria
- Present address:
Max Planck Institute for Multidisciplinary SciencesD‐37077GöttingenGermany
| | - Fatih Demir
- Central Institute for Engineering, Electronics and Analytics52428JülichZEA‐3, Forschungszentrum JülichGermany
- Present address:
Department of BiomedicineAarhus University8000Aarhus CDenmark
| | - Raimund Tenhaken
- Department of Environment and BiodiversityUniversity of Salzburg5020SalzburgAustria
| | - Peter Briza
- Department of Biosciences and Medical BiologyUniversity of Salzburg5020SalzburgAustria
| | - Pitter F. Huesgen
- Central Institute for Engineering, Electronics and Analytics52428JülichZEA‐3, Forschungszentrum JülichGermany
- CECADMedical Faculty and University Hospital, University of Cologne50931CologneGermany
- Institute for Biochemistry, Faculty of Mathematics and Natural SciencesUniversity of Cologne50674CologneGermany
| | - Hans Brandstetter
- Department of Biosciences and Medical BiologyUniversity of Salzburg5020SalzburgAustria
| | - Elfriede Dall
- Department of Biosciences and Medical BiologyUniversity of Salzburg5020SalzburgAustria
| |
Collapse
|
7
|
Holzner C, Böttinger K, Blöchl C, Huber CG, Dahms SO, Dall E, Brandstetter H. Legumain Functions as a Transient TrkB Sheddase. Int J Mol Sci 2023; 24:ijms24065394. [PMID: 36982466 PMCID: PMC10049731 DOI: 10.3390/ijms24065394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
While primarily found in endo-lysosomal compartments, the cysteine protease legumain can also translocate to the cell surface if stabilized by the interaction with the RGD-dependent integrin receptor αVβ3. Previously, it has been shown that legumain expression is inversely related to BDNF-TrkB activity. Here we show that legumain can conversely act on TrkB-BDNF by processing the C-terminal linker region of the TrkB ectodomain in vitro. Importantly, when in complex with BDNF, TrkB was not cleaved by legumain. Legumain-processed TrkB was still able to bind BDNF, suggesting a potential scavenger function of soluble TrkB towards BDNF. The work thus presents another mechanistic link explaining the reciprocal TrkB signaling and δ-secretase activity of legumain, with relevance for neurodegeneration.
Collapse
|
8
|
Wilkinson IVL, Castro-Falcón G, Roda-Serrat MC, Purdy TN, Straetener J, Brauny MM, Maier L, Brötz-Oesterhelt H, Christensen LP, Sieber SA, Hughes CC. The Cyanobacterial "Nutraceutical" Phycocyanobilin Inhibits Cysteine Protease Legumain. Chembiochem 2023; 24:e202200455. [PMID: 36538283 DOI: 10.1002/cbic.202200455] [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: 08/08/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
The blue biliprotein phycocyanin, produced by photo-autotrophic cyanobacteria including spirulina (Arthrospira) and marketed as a natural food supplement or "nutraceutical," is reported to have anti-inflammatory, antioxidant, immunomodulatory, and anticancer activity. These diverse biological activities have been specifically attributed to the phycocyanin chromophore, phycocyanobilin (PCB). However, the mechanism of action of PCB and the molecular targets responsible for the beneficial properties of PCB are not well understood. We have developed a procedure to rapidly cleave the PCB pigment from phycocyanin by ethanolysis and then characterized it as an electrophilic natural product that interacts covalently with thiol nucleophiles but lacks any appreciable cytotoxicity or antibacterial activity against common pathogens and gut microbes. We then designed alkyne-bearing PCB probes for use in chemical proteomics target deconvolution studies. Target identification and validation revealed the cysteine protease legumain (also known as asparaginyl endopeptidase, AEP) to be a target of PCB. Inhibition of this target may account for PCB's diverse reported biological activities.
Collapse
Affiliation(s)
- Isabel V L Wilkinson
- Center for Protein Assemblies (CPA), Department of Chemistry, Technical University of Munich, Ernst-Otto-Fischer-Str. 8, 85748, Garching, Germany
| | - Gabriel Castro-Falcón
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego, CA 92093, USA
| | - Maria C Roda-Serrat
- Department of Green Technology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Trevor N Purdy
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego, CA 92093, USA
| | - Jan Straetener
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076, Tübingen, Germany
| | - Melanie M Brauny
- Cluster of Excellence EXC 2124, Controlling Microbes to Fight Infection, University of Tübingen, 72076, Tübingen, Germany
- Microbiome-Host-Interaction Lab, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076, Tübingen, Germany
| | - Lisa Maier
- Cluster of Excellence EXC 2124, Controlling Microbes to Fight Infection, University of Tübingen, 72076, Tübingen, Germany
- Microbiome-Host-Interaction Lab, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076, Tübingen, Germany
| | - Heike Brötz-Oesterhelt
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076, Tübingen, Germany
- Cluster of Excellence EXC 2124, Controlling Microbes to Fight Infection, University of Tübingen, 72076, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, 72076, Tübingen, Germany
| | - Lars P Christensen
- Department of Green Technology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Stephan A Sieber
- Center for Protein Assemblies (CPA), Department of Chemistry, Technical University of Munich, Ernst-Otto-Fischer-Str. 8, 85748, Garching, Germany
| | - Chambers C Hughes
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego, CA 92093, USA
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076, Tübingen, Germany
- Cluster of Excellence EXC 2124, Controlling Microbes to Fight Infection, University of Tübingen, 72076, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, 72076, Tübingen, Germany
| |
Collapse
|
9
|
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: 25] [Impact Index Per Article: 8.3] [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
|
10
|
Elamin T, Brandstetter H, Dall E. Legumain Activity Is Controlled by Extended Active Site Residues and Substrate Conformation. Int J Mol Sci 2022; 23:12548. [PMID: 36293424 PMCID: PMC9604545 DOI: 10.3390/ijms232012548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Legumain is a lysosomal cysteine protease with strict specificity for cleaving after asparagine residues. By sequence comparison, legumain belongs to MEROPS clan CD of the cysteine proteases, which indicates its structural and mechanistic relation to caspases. Contrasting caspases, legumain harbors a pH-dependent ligase activity in addition to the protease activity. Although we already have a significant body of knowledge on the catalytic activities of legumain, many mechanistic details are still elusive. In this study, we provide evidence that extended active site residues and substrate conformation are steering legumain activities. Biochemical experiments and bioinformatics analysis showed that the catalytic Cys189 and His148 residues are regulated by sterically close Glu190, Ser215 and Asn42 residues. While Glu190 serves as an activity brake, Ser215 and Asn42 have a favorable effect on legumain protease activity. Mutagenesis studies using caspase-9 as model enzyme additionally showed that a similar Glu190 activity brake is also implemented in the caspases. Furthermore, we show that the substrate's conformational flexibility determines whether it will be hydrolyzed or ligated by legumain. The functional understanding of the extended active site residues and of substrate prerequisites will allow us to engineer proteases with increased enzymatic activity and better ligase substrates, with relevance for biotechnological applications.
Collapse
Affiliation(s)
| | | | - Elfriede Dall
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
| |
Collapse
|
11
|
Elamin T, Santos NP, Briza P, Brandstetter H, Dall E. Structural and functional studies of legumain-mycocypin complexes revealed a competitive, exosite-regulated mode of interaction. J Biol Chem 2022; 298:102502. [PMID: 36116553 PMCID: PMC9579014 DOI: 10.1016/j.jbc.2022.102502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/10/2022] [Accepted: 09/12/2022] [Indexed: 12/01/2022] Open
Abstract
Under pathophysiologic conditions such as Alzheimer's disease and cancer, the endolysosomal cysteine protease legumain was found to translocate to the cytosol, the nucleus, and the extracellular space. These noncanonical localizations demand for a tight regulation of legumain activity, which is in part conferred by protein inhibitors. While there is a significant body of knowledge on the interaction of human legumain with endogenous cystatins, only little is known on its regulation by fungal mycocypins. Mycocypins are characterized by (i) versatile, plastic surface loops allowing them to inhibit different classes of enzymes and (ii) a high resistance toward extremes of pH and temperature. These properties make mycocypins attractive starting points for biotechnological and medical applications. In this study, we show that mycocypins utilize an adaptable reactive center loop to target the active site of legumain in a substrate-like manner. The interaction was further stabilized by variable, isoform-specific exosites, converting the substrate recognition into inhibition. Additionally, we found that selected mycocypins were capable of covalent complex formation with legumain by forming a disulfide bond to the active site cysteine. Furthermore, our inhibition studies with other clan CD proteases suggested that mycocypins may serve as broad-spectrum inhibitors of clan CD proteases. Our studies uncovered the potential of mycocypins as a new scaffold for drug development, providing the basis for the design of specific legumain inhibitors.
Collapse
Affiliation(s)
- Tasneem Elamin
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - Naiá P Santos
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - Peter Briza
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - Hans Brandstetter
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - Elfriede Dall
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria.
| |
Collapse
|
12
|
The Asparaginyl Endopeptidase Legumain: An Emerging Therapeutic Target and Potential Biomarker for Alzheimer’s Disease. Int J Mol Sci 2022; 23:ijms231810223. [PMID: 36142134 PMCID: PMC9499314 DOI: 10.3390/ijms231810223] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/21/2022] Open
Abstract
Alzheimer’s disease (AD) is incurable dementia closely associated with aging. Most cases of AD are sporadic, and very few are inherited; the pathogenesis of sporadic AD is complex and remains to be elucidated. The asparaginyl endopeptidase (AEP) or legumain is the only recognized cysteine protease that specifically hydrolyzes peptide bonds after asparagine residues in mammals. The expression level of AEPs in healthy brains is far lower than that of peripheral organs. Recently, growing evidence has indicated that aging may upregulate and overactivate brain AEPs. The overactivation of AEPs drives the onset of AD through cleaving tau and amyloid precursor proteins (APP), and SET, an inhibitor of protein phosphatase 2A (PP2A). The AEP-mediated cleavage of these peptides enhances amyloidosis, promotes tau hyperphosphorylation, and ultimately induces neurodegeneration and cognitive impairment. Upregulated AEPs and related deleterious reactions constitute upstream events of amyloid/tau toxicity in the brain, and represent early pathological changes in AD. Thus, upregulated AEPs are an emerging drug target for disease modification and a potential biomarker for predicting preclinical AD. However, the presence of the blood–brain barrier greatly hinders establishing body-fluid-based methods to measure brain AEPs. Research on AEP-activity-based imaging probes and our recent work suggest that the live brain imaging of AEPs could be used to evaluate its predictive efficacy as an AD biomarker. To advance translational research in this area, AEP imaging probes applicable to human brain and AEP inhibitors with good druggability are urgently needed.
Collapse
|
13
|
Morgan HE, Turnbull WB, Webb ME. Challenges in the use of sortase and other peptide ligases for site-specific protein modification. Chem Soc Rev 2022; 51:4121-4145. [PMID: 35510539 PMCID: PMC9126251 DOI: 10.1039/d0cs01148g] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Site-specific protein modification is a widely-used biochemical tool. However, there are many challenges associated with the development of protein modification techniques, in particular, achieving site-specificity, reaction efficiency and versatility. The engineering of peptide ligases and their substrates has been used to address these challenges. This review will focus on sortase, peptidyl asparaginyl ligases (PALs) and variants of subtilisin; detailing how their inherent specificity has been utilised for site-specific protein modification. The review will explore how the engineering of these enzymes and substrates has led to increased reaction efficiency mainly due to enhanced catalytic activity and reduction of reversibility. It will also describe how engineering peptide ligases to broaden their substrate scope is opening up new opportunities to expand the biochemical toolkit, particularly through the development of techniques to conjugate multiple substrates site-specifically onto a protein using orthogonal peptide ligases. We highlight chemical and biochemical strategies taken to optimise peptide and protein modification using peptide ligases.![]()
Collapse
Affiliation(s)
- Holly E Morgan
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK.
| | - W Bruce Turnbull
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK.
| | - Michael E Webb
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK.
| |
Collapse
|
14
|
Kovalyova Y, Bak DW, Gordon EM, Fung C, Shuman JHB, Cover TL, Amieva MR, Weerapana E, Hatzios SK. An infection-induced oxidation site regulates legumain processing and tumor growth. Nat Chem Biol 2022; 18:698-705. [PMID: 35332331 PMCID: PMC9246868 DOI: 10.1038/s41589-022-00992-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 02/08/2022] [Indexed: 11/15/2022]
Abstract
Oxidative stress is a defining feature of most cancers, including those that stem from carcinogenic infections1. Reactive oxygen species (ROS) can drive tumor formation2–4, yet the molecular oxidation events that contribute to tumorigenesis are largely unknown. Here we show that inactivation of a single, redox-sensitive cysteine in the host protease legumain, which is oxidized during infection with the gastric cancer-causing bacterium Helicobacter pylori, accelerates tumor growth. By using chemical proteomics to map cysteine reactivity in human gastric cells, we determined that H. pylori infection induces oxidation of legumain at Cys219. Legumain oxidation dysregulates intracellular legumain processing and decreases the activity of the enzyme in H. pylori-infected cells. We further show that the site-specific loss of Cys219 reactivity increases tumor growth and mortality in a xenograft model. Our findings establish a link between an infection-induced oxidation site and tumorigenesis while underscoring the importance of cysteine reactivity in tumor growth.
Collapse
Affiliation(s)
- Yekaterina Kovalyova
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA.,Microbial Sciences Institute, Yale University, West Haven, CT, USA.,Department of Chemistry, Yale University, New Haven, CT, USA
| | - Daniel W Bak
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Elizabeth M Gordon
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA.,Microbial Sciences Institute, Yale University, West Haven, CT, USA
| | - Connie Fung
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer H B Shuman
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Timothy L Cover
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA.,Department of Medicine, Vanderbilt University School of Medicine, and Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Manuel R Amieva
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Pediatrics, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Stavroula K Hatzios
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA. .,Microbial Sciences Institute, Yale University, West Haven, CT, USA. .,Department of Chemistry, Yale University, New Haven, CT, USA.
| |
Collapse
|
15
|
Zhang D, Wang Z, Hu S, Chan NY, Liew HT, Lescar J, Tam JP, Liu CF. Asparaginyl Endopeptidase-Mediated Protein C-Terminal Hydrazinolysis for the Synthesis of Bioconjugates. Bioconjug Chem 2022; 33:238-247. [PMID: 34985285 DOI: 10.1021/acs.bioconjchem.1c00551] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Asparaginyl endopeptidases (AEPs) are cysteinyl enzymes naturally catalyzing the hydrolysis and transpeptidation reactions at Asx-Xaa bonds. These reactions go by a common acyl-enzyme thioester intermediate, which is either attacked by water (for a protease-AEP) or by a peptidic amine nucleophile (for a ligase-AEP) to form the respective hydrolysis or aminolysis product. Herein, we show that hydrazine and hydroxylamine, two α-effect nucleophiles, are capable of resolving the thioester intermediate to yield peptide and protein products containing a C-terminal hydrazide and hydroxamic acid functionality, respectively. The hydrazinolysis reaction exhibits very high efficiency and can be completed in minutes at a low enzyme-to-substrate ratio. We further show the utility of the so-formed asparaginyl hydrazide in native chemical ligation and hydrazone conjugation. Using an EGFR-targeting affibody as a model protein, we have showcased our methodology in the preparation of a number of protein ligation or conjugation products, which are decorated with various functional moieties. The ZEGFR affibody-doxorubicin conjugate shows high selective binding and cytotoxicity toward the EGFR-positive A431 cells. Our results demonstrate the advantages of AEP-mediated protein hydrazinolysis as a simple and straightforward strategy for the precision manufacturing of protein bioconjugates.
Collapse
Affiliation(s)
- Dingpeng Zhang
- School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore
| | - Zhen Wang
- School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore
| | - Side Hu
- School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore
| | - Ning-Yu Chan
- School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore
| | - Heng Tai Liew
- School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore
| | - Julien Lescar
- School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore
| | - James P Tam
- School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore
| | - Chuan-Fa Liu
- School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore
| |
Collapse
|
16
|
Liew HT, To J, Zhang X, Hemu X, Chan NY, Serra A, Sze SK, Liu CF, Tam JP. The legumain McPAL1 from Momordica cochinchinensis is a highly stable Asx-specific splicing enzyme. J Biol Chem 2021; 297:101325. [PMID: 34710371 PMCID: PMC8600085 DOI: 10.1016/j.jbc.2021.101325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/09/2021] [Accepted: 10/11/2021] [Indexed: 11/29/2022] Open
Abstract
Legumains, also known as asparaginyl endopeptidases (AEPs), cleave peptide bonds after Asn/Asp (Asx) residues. In plants, certain legumains also have ligase activity that catalyzes biosynthesis of Asx-containing cyclic peptides. An example is the biosynthesis of MCoTI-I/II, a squash family-derived cyclic trypsin inhibitor, which involves splicing to remove the N-terminal prodomain and then N-to-C-terminal cyclization of the mature domain. To identify plant legumains responsible for the maturation of these cyclic peptides, we have isolated and characterized a legumain involved in splicing, McPAL1, from Momordica cochinchinensis (Cucurbitaceae) seeds. Functional studies show that recombinantly expressed McPAL1 displays a pH-dependent, trimodal enzymatic profile. At pH 4 to 6, McPAL1 selectively catalyzed Asp-ligation and Asn-cleavage, but at pH 6.5 to 8, Asn-ligation predominated. With peptide substrates containing N-terminal Asn and C-terminal Asp, such as is found in precursors of MCoTI-I/II, McPAL1 mediates proteolysis at the Asn site and then ligation at the Asp site at pH 5 to 6. Also, McPAL1 is an unusually stable legumain that is tolerant of heat and high pH. Together, our results support that McPAL1 is a splicing legumain at acidic pH that can mediate biosynthesis of MCoTI-I/II. We purport that the high thermal and pH stability of McPAL1 could have applications for protein engineering.
Collapse
Affiliation(s)
- Heng Tai Liew
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Janet To
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Xiaohong Zhang
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Xinya Hemu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Ning-Yu Chan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Aida Serra
- IMDEA Food Research Institute, +Pec Proteomics, Campus of International Excellence UAM+CSIC, Old Cantoblanco Hospital, Cantoblanco, Madrid, Spain; Proteored - Instituto de Salud Carlos III (ISCIII), Campus UAM, Cantoblanco, Madrid, Spain
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Chuan-Fa Liu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - James P Tam
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
17
|
Dall E, Stanojlovic V, Demir F, Briza P, Dahms SO, Huesgen PF, Cabrele C, Brandstetter H. The Peptide Ligase Activity of Human Legumain Depends on Fold Stabilization and Balanced Substrate Affinities. ACS Catal 2021; 11:11885-11896. [PMID: 34621593 PMCID: PMC8491156 DOI: 10.1021/acscatal.1c02057] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/30/2021] [Indexed: 12/11/2022]
Abstract
Protein modification by enzymatic breaking and forming of peptide bonds significantly expands the repertoire of genetically encoded protein sequences. The dual protease-ligase legumain exerts the two opposing activities within a single protein scaffold. Primarily localized to the endolysosomal system, legumain represents a key enzyme in the generation of antigenic peptides for subsequent presentation on the MHCII complex. Here we show that human legumain catalyzes the ligation and cyclization of linear peptides at near-neutral pH conditions, where legumain is intrinsically unstable. Conformational stabilization significantly enhanced legumain's ligase activity, which further benefited from engineering the prime substrate recognition sites for improved affinity. Additionally, we provide evidence that specific legumain activation states allow for differential regulation of its activities. Together these results set the basis for engineering legumain proteases and ligases with applications in biotechnology and drug development.
Collapse
Affiliation(s)
- Elfriede Dall
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Vesna Stanojlovic
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Fatih Demir
- Central Institute for Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Peter Briza
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Sven O. Dahms
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Pitter F. Huesgen
- Central Institute for Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, 52428 Jülich, Germany
- CECAD, Medical Faculty and University Hospital, University of Cologne, 50931 Cologne, Germany
- Institute for Biochemistry, Faculty of Mathematics and Natural Sciences, University of Cologne, 50674 Cologne, Germany
| | - Chiara Cabrele
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Hans Brandstetter
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| |
Collapse
|
18
|
Zhang D, Wang Z, Hu S, Balamkundu S, To J, Zhang X, Lescar J, Tam JP, Liu CF. pH-Controlled Protein Orthogonal Ligation Using Asparaginyl Peptide Ligases. J Am Chem Soc 2021; 143:8704-8712. [PMID: 34096285 DOI: 10.1021/jacs.1c02638] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Peptide asparaginyl ligases (PALs) catalyze transpeptidation at the Asn residue of a short Asn-Xaa1-Xaa2 tripeptide motif. Due to their high catalytic activity toward the P1-Asn substrates at around neutral pH, PALs have been used extensively for peptide ligation at asparaginyl junctions. PALs also bind to aspartyl substrates, but only when the γCOOH of P1-Asp remains in its neutral, protonated form, which usually requires an acidic pH. However, this limits the availability of the amine nucleophile and, consequently, the ligation efficiency at aspartyl junctions. Because of this perceived inefficiency, the use of PALs for Asp-specific ligation remains largely unexplored. We found that PAL enzymes, such as VyPAL2, display appreciable catalytic activities toward P1-Asp substrates at pH 4-5, which are at least 2 orders of magnitude higher than that of sortase A, making them practically useful for both intra- and intermolecular ligations. This also allows sequential ligations, first at Asp and then at Asn junctions, because the newly formed aspartyl peptide bond is resistant to the ligase at the pH used for asparaginyl ligation in the second step. Using this pH-controlled orthogonal ligation method, we dually labeled truncated sfGFP with a cancer-targeting peptide and a doxorubicin derivative at the respective N- and C-terminal ends in the N-to-C direction. In addition, a fluorescein tag and doxorubicin derivative were tagged to an EGFR-targeting affibody in the C-to-N direction. This study shows that the pH-dependent catalytic activity of PAL enzymes can be exploited to prepare multifunction protein biologics for pharmacological applications.
Collapse
Affiliation(s)
- Dingpeng Zhang
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Zhen Wang
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Side Hu
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | | | - Janet To
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Xiaohong Zhang
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Julien Lescar
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - James P Tam
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Chuan-Fa Liu
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| |
Collapse
|
19
|
Zhao J, Fan R, Jia F, Huang Y, Huang Z, Hou Y, Hu SQ. Enzymatic Properties of Recombinant Ligase Butelase-1 and Its Application in Cyclizing Food-Derived Angiotensin I-Converting Enzyme Inhibitory Peptides. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:5976-5985. [PMID: 34003638 DOI: 10.1021/acs.jafc.1c01755] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Butelase-1 is an efficient ligase from Clitoria ternatea with wide applications in the food and biopharmaceutical fields. This research aimed to achieve high-efficiency expression of butelase-1 and explore its application in food-derived angiotensin I-converting enzyme (ACE) inhibitory peptides. The recombinant butelase-1 zymogen was prepared at a yield of 100 mg/L in Escherichia coli and successfully activated at pH 4.5, resulting in a 6973.8 U/L yield of activated butelase-1 with a specific activity of 348.69 U/mg and a catalytic efficiency of 9956 M-1 s-1. Activated butelase-1 exhibited considerable resistance to Tween-20, Triton X-100, and methanol. The "traceless" cyclization of ACE inhibitory peptides was realized using activated butelase-1, which resulted in higher stability and ACE inhibitory activity than those of the linear peptides. Our work proposed an efficient method for the preparation of butelase-1 and provided a promising model for its application in food fields.
Collapse
Affiliation(s)
- Jinsong Zhao
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Renshui Fan
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Feng Jia
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Yanbo Huang
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou, Guangdong 510640, China
| | - Zhiqiang Huang
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Yi Hou
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou, Guangdong 510640, China
| | - Song-Qing Hu
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
20
|
Zhang W, Lin Y. The Mechanism of Asparagine Endopeptidase in the Progression of Malignant Tumors: A Review. Cells 2021; 10:cells10051153. [PMID: 34068767 PMCID: PMC8151911 DOI: 10.3390/cells10051153] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/24/2021] [Accepted: 05/07/2021] [Indexed: 12/20/2022] Open
Abstract
Asparagine endopeptidase (AEP), also called legumain, is currently the only known cysteine protease that specifically cleaves peptide bonds in asparaginyl residue in the mammalian genome. Since 2003, AEP has been reported to be widely expressed in a variety of carcinomas and is considered a potential therapeutic target. In the following years, researchers intensively investigated the substrates of AEP and the mechanism of AEP in partial tumors. With the identification of substrate proteins such as P53, integrin αvβ3, MMP-2, and MMP-9, the biochemical mechanism of AEP in carcinomas is also more precise. This review will clarify the probable mechanisms of AEP in the progression of breast carcinoma, glioblastoma, gastric carcinoma, and epithelial ovarian carcinoma. This review will also discuss the feasibility of targeted therapy with AEP inhibitor (AEPI) in these carcinomas.
Collapse
|
21
|
Nonis SG, Haywood J, Mylne JS. Plant asparaginyl endopeptidases and their structural determinants of function. Biochem Soc Trans 2021; 49:965-976. [PMID: 33666219 PMCID: PMC8106488 DOI: 10.1042/bst20200908] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/14/2022]
Abstract
Asparaginyl endopeptidases (AEPs) are versatile enzymes that in biological systems are involved in producing three different catalytic outcomes for proteins, namely (i) routine cleavage by bond hydrolysis, (ii) peptide maturation, including macrocyclisation by a cleavage-coupled intramolecular transpeptidation and (iii) circular permutation involving separate cleavage and transpeptidation reactions resulting in a major reshuffling of protein sequence. AEPs differ in their preference for cleavage or transpeptidation reactions, catalytic efficiency, and preference for asparagine or aspartate target residues. We look at structural analyses of various AEPs that have laid the groundwork for identifying important determinants of AEP function in recent years, with much of the research impetus arising from the potential biotechnological and pharmaceutical applications.
Collapse
Affiliation(s)
- Samuel G. Nonis
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, Perth 6009, Australia
- The ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, 35 Stirling Highway, Crawley, Perth 6009, Australia
| | - Joel Haywood
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, Perth 6009, Australia
- The ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, 35 Stirling Highway, Crawley, Perth 6009, Australia
| | - Joshua S. Mylne
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, Perth 6009, Australia
- The ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, 35 Stirling Highway, Crawley, Perth 6009, Australia
| |
Collapse
|
22
|
Hemu X, Zhang X, Nguyen GKT, To J, Serra A, Loo S, Sze SK, Liu CF, Tam JP. Characterization and application of natural and recombinant butelase-1 to improve industrial enzymes by end-to-end circularization. RSC Adv 2021; 11:23105-23112. [PMID: 35480425 PMCID: PMC9034278 DOI: 10.1039/d1ra03763c] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/23/2021] [Indexed: 01/14/2023] Open
Abstract
Butelase-1, an asparaginyl endopeptidase or legumain, is the prototypical and fastest known Asn/Asp-specific peptide ligase that could be used for improving other enzymes by catalyzing simple and efficient end-to-end circularization.
Collapse
Affiliation(s)
- Xinya Hemu
- School of Biological Sciences
- Nanyang Technological University
- Singapore
| | - Xiaohong Zhang
- School of Biological Sciences
- Nanyang Technological University
- Singapore
| | - Giang K. T. Nguyen
- WIL@NUS Corporate Lab
- MD6 Centre for Translational Medicine
- Wilmar International Limited
- National University of Singapore
- Singapore
| | - Janet To
- School of Biological Sciences
- Nanyang Technological University
- Singapore
| | - Aida Serra
- IMDEA Food Research Institute
- +Pec Proteomics
- Campus of International Excellence UAM+CSIC
- Old Cantoblanco Hospital
- Madrid 28049
| | - Shining Loo
- School of Biological Sciences
- Nanyang Technological University
- Singapore
| | - Siu Kwan Sze
- School of Biological Sciences
- Nanyang Technological University
- Singapore
| | - Chuan-Fa Liu
- School of Biological Sciences
- Nanyang Technological University
- Singapore
| | - James P. Tam
- School of Biological Sciences
- Nanyang Technological University
- Singapore
| |
Collapse
|
23
|
Legumain Induces Oral Cancer Pain by Biased Agonism of Protease-Activated Receptor-2. J Neurosci 2020; 41:193-210. [PMID: 33172978 DOI: 10.1523/jneurosci.1211-20.2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most painful cancers, which interferes with orofacial function including talking and eating. We report that legumain (Lgmn) cleaves protease-activated receptor-2 (PAR2) in the acidic OSCC microenvironment to cause pain. Lgmn is a cysteine protease of late endosomes and lysosomes that can be secreted; it exhibits maximal activity in acidic environments. The role of Lgmn in PAR2-dependent cancer pain is unknown. We studied Lgmn activation in human oral cancers and oral cancer mouse models. Lgmn was activated in OSCC patient tumors, compared with matched normal oral tissue. After intraplantar, facial or lingual injection, Lgmn evoked nociception in wild-type (WT) female mice but not in female mice lacking PAR2 in NaV1.8-positive neurons (Par2Nav1.8), nor in female mice treated with a Lgmn inhibitor, LI-1. Inoculation of an OSCC cell line caused mechanical and thermal hyperalgesia that was reversed by LI-1. Par2Nav1.8 and Lgmn deletion attenuated mechanical allodynia in female mice with carcinogen-induced OSCC. Lgmn caused PAR2-dependent hyperexcitability of trigeminal neurons from WT female mice. Par2 deletion, LI-1, and inhibitors of adenylyl cyclase or protein kinase A (PKA) prevented the effects of Lgmn. Under acidified conditions, Lgmn cleaved within the extracellular N terminus of PAR2 at Asn30↓Arg31, proximal to the canonical trypsin activation site. Lgmn activated PAR2 by biased mechanisms in HEK293 cells to induce Ca2+ mobilization, cAMP formation, and PKA/protein kinase D (PKD) activation, but not β-arrestin recruitment or PAR2 endocytosis. Thus, in the acidified OSCC microenvironment, Lgmn activates PAR2 by biased mechanisms that evoke cancer pain.SIGNIFICANCE STATEMENT Oral squamous cell carcinoma (OSCC) is one of the most painful cancers. We report that legumain (Lgmn), which exhibits maximal activity in acidic environments, cleaves protease-activated receptor-2 (PAR2) on neurons to produce OSCC pain. Active Lgmn was elevated in OSCC patient tumors, compared with matched normal oral tissue. Lgmn evokes pain-like behavior through PAR2 Exposure of pain-sensing neurons to Lgmn decreased the current required to generate an action potential through PAR2 Inhibitors of adenylyl cyclase and protein kinase A (PKA) prevented the effects of Lgmn. Lgmn activated PAR2 to induce calcium mobilization, cAMP formation, and activation of protein kinase D (PKD) and PKA, but not β-arrestin recruitment or PAR2 endocytosis. Thus, Lgmn is a biased agonist of PAR2 that evokes cancer pain.
Collapse
|
24
|
Dall E, Zauner FB, Soh WT, Demir F, Dahms SO, Cabrele C, Huesgen PF, Brandstetter H. Structural and functional studies of Arabidopsis thaliana legumain beta reveal isoform specific mechanisms of activation and substrate recognition. J Biol Chem 2020; 295:13047-13064. [PMID: 32719006 PMCID: PMC7489914 DOI: 10.1074/jbc.ra120.014478] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/14/2020] [Indexed: 01/19/2023] Open
Abstract
The vacuolar cysteine protease legumain plays important functions in seed maturation and plant programmed cell death. Because of their dual protease and ligase activity, plant legumains have become of particular biotechnological interest, e.g. for the synthesis of cyclic peptides for drug design or for protein engineering. However, the molecular mechanisms behind their dual protease and ligase activities are still poorly understood, limiting their applications. Here, we present the crystal structure of Arabidopsis thaliana legumain isoform β (AtLEGβ) in its zymogen state. Combining structural and biochemical experiments, we show for the first time that plant legumains encode distinct, isoform-specific activation mechanisms. Whereas the autocatalytic activation of isoform γ (AtLEGγ) is controlled by the latency-conferring dimer state, the activation of the monomeric AtLEGβ is concentration independent. Additionally, in AtLEGβ the plant-characteristic two-chain intermediate state is stabilized by hydrophobic rather than ionic interactions, as in AtLEGγ, resulting in significantly different pH stability profiles. The crystal structure of AtLEGβ revealed unrestricted nonprime substrate binding pockets, consistent with the broad substrate specificity, as determined by degradomic assays. Further to its protease activity, we show that AtLEGβ exhibits a true peptide ligase activity. Whereas cleavage-dependent transpeptidase activity has been reported for other plant legumains, AtLEGβ is the first example of a plant legumain capable of linking free termini. The discovery of these isoform-specific differences will allow us to identify and rationally design efficient ligases with application in biotechnology and drug development.
Collapse
Affiliation(s)
- Elfriede Dall
- Department of Biosciences, University of Salzburg, Salzburg, Austria.
| | - Florian B Zauner
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Wai Tuck Soh
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Fatih Demir
- Central Institute for Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany
| | - Sven O Dahms
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Chiara Cabrele
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Pitter F Huesgen
- Central Institute for Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany; CECAD, Medical Faculty and University Hospital, University of Cologne, Cologne, Germany; Institute for Biochemistry, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Hans Brandstetter
- Department of Biosciences, University of Salzburg, Salzburg, Austria.
| |
Collapse
|
25
|
Poreba M. Recent advances in the development of legumain-selective chemical probes and peptide prodrugs. Biol Chem 2020; 400:1529-1550. [PMID: 31021817 DOI: 10.1515/hsz-2019-0135] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022]
Abstract
Legumain, which is also known as vacuolar processing enzyme (VPE) or asparaginyl endopeptidase (AEP), is a cysteine protease that was first discovered and characterized in the leguminous seeds of the moth bean in the early 1990s. Later, this enzyme was also detected in higher organisms, including eukaryotes. This pH-dependent protease displays the highest activity in acidic endolysosomal compartments; however, legumain also displays nuclear, cytosolic and extracellular activity when stabilized by other proteins or intramolecular complexes. Based on the results from over 25 years of research, this protease is involved in multiple cellular events, including protein degradation and antigen presentation. Moreover, when dysregulated, this protease contributes to the progression of several diseases, with cancer being the well-studied example. Research on legumain biology was undoubtedly facilitated by the use of small molecule chemical tools. Therefore, in this review, I present the historical perspectives and most current strategies for the development of small molecule substrates, inhibitors and activity-based probes for legumain. These tools are of paramount importance in elucidating the roles of legumain in multiple biological processes. Finally, as this enzyme appears to be a promising molecular target for anticancer therapies, the development of legumain-activated prodrugs is also described.
Collapse
Affiliation(s)
- Marcin Poreba
- Department of Bioorganic Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370, Wroclaw, Poland
| |
Collapse
|
26
|
Du J, Yap K, Chan LY, Rehm FBH, Looi FY, Poth AG, Gilding EK, Kaas Q, Durek T, Craik DJ. A bifunctional asparaginyl endopeptidase efficiently catalyzes both cleavage and cyclization of cyclic trypsin inhibitors. Nat Commun 2020; 11:1575. [PMID: 32221295 PMCID: PMC7101308 DOI: 10.1038/s41467-020-15418-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 03/06/2020] [Indexed: 01/08/2023] Open
Abstract
Asparaginyl endopeptidases (AEPs) catalyze the key backbone cyclization step during the biosynthesis of plant-derived cyclic peptides. Here, we report the identification of two AEPs from Momordica cochinchinensis and biochemically characterize MCoAEP2 that catalyzes the maturation of trypsin inhibitor cyclotides. Recombinantly produced MCoAEP2 catalyzes the backbone cyclization of a linear cyclotide precursor (MCoTI-II-NAL) with a kcat/Km of 620 mM−1 s−1, making it one of the fastest cyclases reported to date. We show that MCoAEP2 can mediate both the N-terminal excision and C-terminal cyclization of cyclotide precursors in vitro. The rate of cyclization/hydrolysis is primarily influenced by varying pH, which could potentially control the succession of AEP-mediated processing events in vivo. Furthermore, MCoAEP2 efficiently catalyzes the backbone cyclization of an engineered MCoTI-II analog with anti-angiogenic activity. MCoAEP2 provides enhanced synthetic access to structures previously inaccessible by direct chemistry approaches and enables the wider application of trypsin inhibitor cyclotides in biotechnology applications. Asparaginyl endopeptidases (AEPs) catalyze the cyclization step during the biosynthesis of cyclic peptides in plants. Here, the authors report a recombinantly produced AEP that catalyzes the backbone cyclization of a linear cyclotide precursor and an engineered analog with high efficiency and in a pH-dependent manner.
Collapse
Affiliation(s)
- Junqiao Du
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Kuok Yap
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Lai Yue Chan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Fabian B H Rehm
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Fong Yang Looi
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Aaron G Poth
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Edward K Gilding
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Quentin Kaas
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Thomas Durek
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia.
| | - David J Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia.
| |
Collapse
|
27
|
Soh WT, Demir F, Dall E, Perrar A, Dahms SO, Kuppusamy M, Brandstetter H, Huesgen PF. ExteNDing Proteome Coverage with Legumain as a Highly Specific Digestion Protease. Anal Chem 2020; 92:2961-2971. [PMID: 31951383 PMCID: PMC7075662 DOI: 10.1021/acs.analchem.9b03604] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Bottom-up
mass spectrometry-based proteomics utilizes proteolytic
enzymes with well characterized specificities to generate peptides
amenable for identification by high-throughput tandem mass spectrometry.
Trypsin, which cuts specifically after the basic residues lysine and
arginine, is the predominant enzyme used for proteome digestion, although
proteases with alternative specificities are required to detect sequences
that are not accessible after tryptic digest. Here, we show that the
human cysteine protease legumain exhibits a strict substrate specificity
for cleavage after asparagine and aspartic acid residues during in-solution
digestions of proteomes extracted from Escherichia
coli, mouse embryonic fibroblast cell cultures, and Arabidopsis thaliana leaves. Generating peptides
highly complementary in sequence, yet similar in their biophysical
properties, legumain (as compared to trypsin or GluC) enabled complementary
proteome and protein sequence coverage. Importantly, legumain further
enabled the identification and enrichment of protein N-termini not
accessible in GluC- or trypsin-digested samples. Legumain cannot cleave
after glycosylated Asn residues, which enabled the robust identification
and orthogonal validation of N-glycosylation sites based on alternating
sequential sample treatments with legumain and PNGaseF and vice versa.
Taken together, we demonstrate that legumain is a practical, efficient
protease for extending the proteome and sequence coverage achieved
with trypsin, with unique possibilities for the characterization of
post-translational modification sites.
Collapse
Affiliation(s)
- Wai Tuck Soh
- Department of Biosciences , University of Salzburg , 5020 Salzburg , Austria
| | - Fatih Demir
- Central Institute for Engineering, Electronics and Analytics, ZEA-3 , Forschungszentrum Jülich , 52428 Jülich , Germany
| | - Elfriede Dall
- Department of Biosciences , University of Salzburg , 5020 Salzburg , Austria
| | - Andreas Perrar
- Central Institute for Engineering, Electronics and Analytics, ZEA-3 , Forschungszentrum Jülich , 52428 Jülich , Germany
| | - Sven O Dahms
- Department of Biosciences , University of Salzburg , 5020 Salzburg , Austria
| | - Maithreyan Kuppusamy
- Central Institute for Engineering, Electronics and Analytics, ZEA-3 , Forschungszentrum Jülich , 52428 Jülich , Germany
| | - Hans Brandstetter
- Department of Biosciences , University of Salzburg , 5020 Salzburg , Austria
| | - Pitter F Huesgen
- Central Institute for Engineering, Electronics and Analytics, ZEA-3 , Forschungszentrum Jülich , 52428 Jülich , Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, Medical Faculty and University Hospital , University of Cologne , 50931 Cologne , Germany.,Institute for Biochemistry, Faculty of Mathematics and Natural Sciences , University of Cologne , 50674 Cologne , Germany
| |
Collapse
|
28
|
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: 46] [Impact Index Per Article: 9.2] [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
|
29
|
Progress toward sourcing plants for new bioconjugation tools: a screening evaluation of a model peptide ligase using a synthetic precursor. 3 Biotech 2019; 9:442. [PMID: 31763120 DOI: 10.1007/s13205-019-1983-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/30/2019] [Indexed: 10/25/2022] Open
Abstract
In the present study, leaves from 39 phylogenetically distant plant species were sampled and screened for asparaginyl endopeptidase ligase activity using mass spectrometry to test the generality of peptide ligases in plants. A modified version of the sunflower trypsin inhibitor-1 precursor was used as the substrate for reactions with leaf crude extracts and protein fractions. Masses consistent with products of asparaginyl endopeptidase activities that cleave and ligate the substrate into cyclic peptide following the reactions were detected in 8 plants: Nerium oleander and Thevetia peruviana of the family Apocynaceae; Bauhinia variegata, Dermatophyllum secundiflorum, Pithecellobium flexicaule, and Prosopis chilensis of the family Fabaceae; Morus alba of the family Moraceae; and Citrus aurantium of the family Rutaceae. This screening result represents a 20% hit rate for finding asparaginyl endopeptidase ligase activity from the arbitrary plants sampled. Analysis following a 2-h reaction of the substrate with the crude extract of D. secundiflorum leaves showed that the yield of cyclic peptide remained stable around 0.5 ± 0.1% of the substrate over the course of the reaction.
Collapse
|
30
|
Umei TC, Kishimoto Y, Aoyama M, Saita E, Niki H, Ikegami Y, Ohmori R, Kondo K, Momiyama Y. High Plasma Levels of Legumain in Patients with Complex Coronary Lesions. J Atheroscler Thromb 2019; 27:711-717. [PMID: 31735728 PMCID: PMC7406406 DOI: 10.5551/jat.52027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Aim: The degradation of the vascular extracellular matrix is important for atherosclerosis. The cysteine protease legumain was shown to be upregulated in atherosclerotic plaques, especially unstable plaques. However, no study has reported blood legumain levels in patients with coronary artery disease (CAD). Methods: We investigated plasma legumain and C-reactive protein (CRP) levels in 372 patients undergoing elective coronary angiography. Results: CAD was found in 225 patients. Compared with patients without CAD, those with CAD had higher CRP levels (median 0.60 [0.32, 1.53] vs. 0.46 [0.22, 0.89] mg/L, P < 0.001), but no difference was found in legumain levels between patients with and without CAD (median 5.08 [3.87, 6.82] vs. 4.99 [3.84, 6.88] ng/mL). A stepwise increase in CRP was found depending on the number of > 50% stenotic vessels: 0.55 mg/L in 1-vessel, 0.71 mg/L in 2-vessel, and 0.86 mg/L in 3-vessel diseases (P < 0.001). However, legumain did not differ among 1-, 2-, and 3-vessel diseases (5.20, 4.93, and 5.01 ng/mL, respectively). Of 225 patients with CAD, 40 (18%) had complex lesions. No difference was found in CRP levels between patients with CAD with and without complex lesions (0.60 [0.34, 1.53] vs. 0.60 [0.32, 1.51] mg/L). Notably, legumain levels were higher in patients with CAD with complex lesions than without such lesions (6.05 [4.64, 8.64] vs. 4.93 [3.76, 6.52] ng/mL, P < 0.01). In multivariate analysis, legumain levels were not a factor for CAD, but were a factor for complex lesions. The odds ratio for complex lesions was 2.45 (95% CI = 1.26–4.79) for legumain > 5.5 ng/mL. Conclusion: Plasma legumain levels were associated with the presence of complex coronary lesions.
Collapse
Affiliation(s)
- Tomohiko C Umei
- Department of Cardiology, National Hospital Organization Tokyo Medical Center
| | - Yoshimi Kishimoto
- Endowed Research Department "Food for Health", Ochanomizu University
| | - Masayuki Aoyama
- Department of Cardiology, National Hospital Organization Tokyo Medical Center
| | - Emi Saita
- Endowed Research Department "Food for Health", Ochanomizu University
| | - Hanako Niki
- Department of Cardiology, National Hospital Organization Tokyo Medical Center
| | - Yukinori Ikegami
- Department of Cardiology, National Hospital Organization Tokyo Medical Center
| | - Reiko Ohmori
- Faculty of Regional Design, Utsunomiya University
| | - Kazuo Kondo
- Endowed Research Department "Food for Health", Ochanomizu University.,Institute of Life Innovation Studies, Toyo University
| | - Yukihiko Momiyama
- Department of Cardiology, National Hospital Organization Tokyo Medical Center
| |
Collapse
|
31
|
Schlegel K, Awwad K, Heym RG, Holzinger D, Doell A, Barghorn S, Jahn TR, Klein C, Mordashova Y, Schulz M, Gasparini L. N368-Tau fragments generated by legumain are detected only in trace amount in the insoluble Tau aggregates isolated from AD brain. Acta Neuropathol Commun 2019; 7:177. [PMID: 31722749 PMCID: PMC6854719 DOI: 10.1186/s40478-019-0831-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 10/17/2019] [Indexed: 01/20/2023] Open
Abstract
Intraneuronal insoluble inclusions made of Tau protein are neuropathological hallmarks of Alzheimer Disease (AD). Cleavage of Tau by legumain (LGMN) has been proposed to be crucial for aggregation of Tau into fibrils. However, it remains unclear if LGMN-cleaved Tau fragments accumulate in AD Tau inclusions.Using an in vitro enzymatic assay and non-targeted mass spectrometry, we identified four putative LGMN cleavage sites at Tau residues N167-, N255-, N296- and N368. Cleavage at N368 generates variously sized N368-Tau fragments that are aggregation prone in the Thioflavin T assay in vitro. N368-cleaved Tau is not detected in the brain of legumain knockout mice, indicating that LGMN is required for Tau cleavage in the mouse brain in vivo. Using a targeted mass spectrometry method in combination with tissue fractionation and biochemical analysis, we investigated whether N368-cleaved Tau is differentially produced and aggregated in brain of AD patients and control subjects. In brain soluble extracts, despite reduced uncleaved Tau in AD, levels of N368-cleaved Tau are comparable in AD and control hippocampus, suggesting that LGMN-mediated cleavage of Tau is not altered in AD. Consistently, levels of activated, cleaved LGMN are also similar in AD and control brain extracts. To assess the potential accumulation of N368-cleaved Tau in insoluble Tau aggregates, we analyzed sarkosyl-insoluble extracts from AD and control hippocampus. Both N368-cleaved Tau and uncleaved Tau were significantly increased in AD as a consequence of pathological Tau inclusions accumulation. However, the amount of N368-cleaved Tau represented only a very minor component (< 0.1%) of insoluble Tau.Our data indicate that LGMN physiologically cleaves Tau in the mouse and human brain generating N368-cleaved Tau fragments, which remain largely soluble and are present only in low proportion in Tau insoluble aggregates compared to uncleaved Tau. This suggests that LGMN-cleaved Tau has limited role in the progressive accumulation of Tau inclusions in AD.
Collapse
|
32
|
Lunde NN, Bosnjak T, Solberg R, Johansen HT. Mammalian legumain – A lysosomal cysteine protease with extracellular functions? Biochimie 2019; 166:77-83. [DOI: 10.1016/j.biochi.2019.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/04/2019] [Indexed: 12/31/2022]
|
33
|
Toss MS, Miligy IM, Gorringe KL, McCaffrey L, AlKawaz A, Abidi A, Ellis IO, Green AR, Rakha EA. Legumain is an independent predictor for invasive recurrence in breast ductal carcinoma in situ. Mod Pathol 2019; 32:639-649. [PMID: 30429518 DOI: 10.1038/s41379-018-0180-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/01/2018] [Accepted: 11/01/2018] [Indexed: 12/21/2022]
Abstract
Legumain is a proteolytic enzyme that plays a role in the regulation of cell proliferation in invasive breast cancer. Studies evaluating its role in ductal carcinoma in situ (DCIS) are lacking. Here, we aimed to characterize legumain protein expression in DCIS and evaluate its prognostic significance. Legumain was assessed immunohistochemically in a tissue microarray of a well-characterized cohort of DCIS (n = 776 pure DCIS and n = 239 DCIS associated with invasive breast cancer (DCIS-mixed)). Legumain immunoreactivity was scored in tumor cells and surrounding stroma and related to clinicopathological parameters and patient outcome. High legumain expression was observed in 23% of pure DCIS and was associated with features of high-risk DCIS including higher nuclear grade, comedo necrosis, hormone receptor negativity, HER2 positivity, and higher proliferation index. Legumain expression was higher in DCIS associated with invasive breast cancer than in pure DCIS (p < 0.0001). In the DCIS-mixed cohort, the invasive component showed higher legumain expression than the DCIS component (p < 0.0001). Legumain was an independent predictor of shorter local recurrencefree interval for all recurrences (p = 0.0003) and for invasive recurrences (p = 0.002). When incorporated with other risk factors, legumain provided better patient risk stratification. High legumain expression is associated with poor prognosis in DCIS and could be a potential marker to predict DCIS progression to invasive disease.
Collapse
Affiliation(s)
- Michael S Toss
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK.,Histopathology department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Islam M Miligy
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK.,Histopathology department, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt
| | - Kylie L Gorringe
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - L McCaffrey
- Department of Oncology, Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Abdulbaqi AlKawaz
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK.,College of dentistry, Al Mustansiriya University, Baghdad, Iraq
| | - Asima Abidi
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Ian O Ellis
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Andrew R Green
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Emad A Rakha
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK. .,Histopathology department, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt.
| |
Collapse
|
34
|
Dahms SO, Demir F, Huesgen PF, Thorn K, Brandstetter H. Sirtilins - the new old members of the vitamin K-dependent coagulation factor family. J Thromb Haemost 2019; 17:470-481. [PMID: 30644641 PMCID: PMC6850207 DOI: 10.1111/jth.14384] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Indexed: 12/04/2022]
Abstract
Essentials Blood coagulation is driven by vitamin K (VK)-dependent proteases. We have identified and characterized 'sirtilin' as an additional VK-dependent protease. Sirtilins emerged early in the evolution of the coagulation system of vertebrates. Ubiquitous occurrence might indicate an important functional role of sirtilins. SUMMARY: Background Vitamin K (VK)-dependent proteases are major players in blood coagulation, including both the initiation and the regulation of the cascade. Five different members of this protease family have been described, comprising the following coagulation factors: factor VII, FIX, FX, protein C (PC), and prothrombin (FII). FVII, FIX, FX and PC share a typical domain architecture, with an N-terminal γ-carboxyglutamate (Gla) domain, two epidermal growth factor-like (EGF) domains, and a C-terminal trypsin-like serine protease (SP) domain. Objectives We have identified uncharacterized proteins in snake genomes showing the typical Gla-EGF1-EGF2-SP domain architecture but relatively low sequence conservation compared to known VK-dependent proteases. On the basis of sequence analysis, we hypothesized that these proteins are functional members of the VK-dependent protease family. Methods/results Using phylogenetic analyses, we confirmed the so-called 'sirtilins' as an additional VK-dependent protease class. These proteases were found in several vertebrates, including jawless fish, cartilaginous fish, bony fish, reptiles, birds, and marsupials, but not in other mammals. The recombinant zymogen form of Thamnophis sirtalis sirtilin was produced by in vitro renaturation, and was activated with human activated FXI. The activated form of sirtilin proteolytically cleaved peptide and protein substrates, including prothrombin. Mass spectrometry-based substrate profiling of sirtilin revealed a narrower sequence specificity than those of FIX and FX. Conclusions The ubiquitous occurrence of sirtilins in many vertebrate classes might indicate an important functional role. Understanding the detailed functions of sirtilins might contribute to a deeper understanding of the evolution and function of the vertebrate coagulation system.
Collapse
Affiliation(s)
- Sven O. Dahms
- Department of BiosciencesUniversity of SalzburgSalzburgAustria
| | - Fatih Demir
- ZEA‐3 AnalyticsCentral Institute for Engineering, Electronics and AnalyticsForschungszentrum JülichJülichGermany
| | - Pitter F. Huesgen
- ZEA‐3 AnalyticsCentral Institute for Engineering, Electronics and AnalyticsForschungszentrum JülichJülichGermany
| | - Karina Thorn
- Haemophilia ResearchNovo Nordisk A/SMåløvDenmark
| | | |
Collapse
|
35
|
Jutras PV, Goulet M, Lavoie P, D'Aoust M, Sainsbury F, Michaud D. Recombinant protein susceptibility to proteolysis in the plant cell secretory pathway is pH-dependent. PLANT BIOTECHNOLOGY JOURNAL 2018; 16:1928-1938. [PMID: 29618167 PMCID: PMC6181212 DOI: 10.1111/pbi.12928] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/14/2018] [Accepted: 03/21/2018] [Indexed: 05/07/2023]
Abstract
Cellular engineering approaches have been proposed to mitigate unintended proteolysis in plant protein biofactories, involving the design of protease activity-depleted environments by gene silencing or in situ inactivation with accessory protease inhibitors. Here, we assessed the impact of influenza virus M2 proton channel on host protease activities and recombinant protein processing in the cell secretory pathway of Nicotiana benthamiana leaves. Transient co-expression assays with M2 and GFP variant pHluorin were first conducted to illustrate the potential of proton export from the Golgi lumen to promote recombinant protein yield. A fusion protein-based system involving protease-sensitive peptide linkers to attach inactive variants of tomato cystatin SlCYS8 was then designed to relate the effects of M2 on protein levels with altered protease activities in situ. Secreted versions of the cystatin fusions transiently expressed in leaf tissue showed variable 'fusion to free cystatin' cleavage ratios, in line with the occurrence of protease forms differentially active against the peptide linkers in the secretory pathway. Variable ratios were also observed for the fusions co-expressed with M2, but the extent of fusion cleavage was changed for several fusions, positively or negatively, as a result of pH increase in the Golgi. These data indicating a remodelling of endogenous protease activities upon M2 expression confirm that the stability of recombinant proteins in the plant cell secretory pathway is pH-dependent. They suggest, in practice, the potential of M2 proton channel to modulate the stability of protease-susceptible secreted proteins in planta via a pH-related, indirect effect on host resident proteases.
Collapse
Affiliation(s)
- Philippe V. Jutras
- Centre de recherche et d'innovation sur les végétauxUniversité LavalQuebec CityQCCanada
| | - Marie‐Claire Goulet
- Centre de recherche et d'innovation sur les végétauxUniversité LavalQuebec CityQCCanada
| | | | | | - Frank Sainsbury
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt LuciaQldAustralia
| | - Dominique Michaud
- Centre de recherche et d'innovation sur les végétauxUniversité LavalQuebec CityQCCanada
| |
Collapse
|
36
|
Dall E, Hollerweger JC, Dahms SO, Cui H, Häussermann K, Brandstetter H. Structural and functional analysis of cystatin E reveals enzymologically relevant dimer and amyloid fibril states. J Biol Chem 2018; 293:13151-13165. [PMID: 29967063 PMCID: PMC6109925 DOI: 10.1074/jbc.ra118.002154] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 06/26/2018] [Indexed: 12/26/2022] Open
Abstract
Protein activity is often regulated by altering the oligomerization state. One mechanism of multimerization involves domain swapping, wherein proteins exchange parts of their structures and thereby form long-lived dimers or multimers. Domain swapping has been specifically observed in amyloidogenic proteins, for example the cystatin superfamily of cysteine protease inhibitors. Cystatins are twin-headed inhibitors, simultaneously targeting the lysosomal cathepsins and legumain, with important roles in cancer progression and Alzheimer's disease. Although cystatin E is the most potent legumain inhibitor identified so far, nothing is known about its propensity to oligomerize. In this study, we show that conformational destabilization of cystatin E leads to the formation of a domain-swapped dimer with increased conformational stability. This dimer was active as a legumain inhibitor by forming a trimeric complex. By contrast, the binding sites toward papain-like proteases were buried within the cystatin E dimer. We also showed that the dimers could further convert to amyloid fibrils. Unexpectedly, cystatin E amyloid fibrils contained functional protein, which inhibited both legumain and papain-like enzymes. Fibril formation was further regulated by glycosylation. We speculate that cystatin amyloid fibrils might serve as a binding platform to stabilize the pH-sensitive legumain and cathepsins in the extracellular environment, contributing to their physiological and pathological functions.
Collapse
Affiliation(s)
- Elfriede Dall
- From the Department of Biosciences, University of Salzburg, A-5020 Salzburg, Austria and
| | - Julia C Hollerweger
- From the Department of Biosciences, University of Salzburg, A-5020 Salzburg, Austria and
| | - Sven O Dahms
- From the Department of Biosciences, University of Salzburg, A-5020 Salzburg, Austria and
| | - Haissi Cui
- the Center for Integrated Protein Science Munich, Technical University of Munich, D-85748 Munich, Germany
| | - Katharina Häussermann
- the Center for Integrated Protein Science Munich, Technical University of Munich, D-85748 Munich, Germany
| | - Hans Brandstetter
- From the Department of Biosciences, University of Salzburg, A-5020 Salzburg, Austria and
| |
Collapse
|
37
|
Zauner FB, Dall E, Regl C, Grassi L, Huber CG, Cabrele C, Brandstetter H. Crystal Structure of Plant Legumain Reveals a Unique Two-Chain State with pH-Dependent Activity Regulation. THE PLANT CELL 2018; 30:686-699. [PMID: 29453229 PMCID: PMC5894848 DOI: 10.1105/tpc.17.00963] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/13/2018] [Accepted: 02/13/2018] [Indexed: 05/10/2023]
Abstract
The vacuolar cysteine protease legumain can cleave and selectively rebuild peptide bonds, thereby vastly expanding the sequential repertoire of biomolecules. In this context, plant legumains have recently attracted particular interest. Furthermore, legumains have important roles in many physiological processes, including programmed cell death. Their efficient peptide bond ligase activity has gained tremendous interest in the design of cyclic peptides for drug design. However, the mechanistic understanding of these dual activities is incomplete and partly conflicting. Here, we present the crystal structure of a plant legumain, Arabidopsis thaliana isoform-γ (AtLEGγ). Employing a conserved legumain fold, the plant legumain AtLEGγ revealed unique mechanisms of autoactivation, including a plant-specific two-chain activation state, which remains conformationally stable at neutral pH, which is a prerequisite for full ligase activity and survival in different cell compartments. The charge distribution around the α6-helix mediates the pH-dependent dimerization and serves as a gatekeeper for the active site, thus regulating its protease and ligase activity.
Collapse
Affiliation(s)
- Florian B Zauner
- Department of Molecular Biology and Christian Doppler Laboratory for Biosimilar Research, University of Salzburg, A-5020 Salzburg, Austria
| | - Elfriede Dall
- Department of Molecular Biology and Christian Doppler Laboratory for Biosimilar Research, University of Salzburg, A-5020 Salzburg, Austria
| | - Christof Regl
- Department of Molecular Biology and Christian Doppler Laboratory for Biosimilar Research, University of Salzburg, A-5020 Salzburg, Austria
| | - Luigi Grassi
- Department of Molecular Biology and Christian Doppler Laboratory for Biosimilar Research, University of Salzburg, A-5020 Salzburg, Austria
| | - Christian G Huber
- Department of Molecular Biology and Christian Doppler Laboratory for Biosimilar Research, University of Salzburg, A-5020 Salzburg, Austria
| | - Chiara Cabrele
- Department of Molecular Biology and Christian Doppler Laboratory for Biosimilar Research, University of Salzburg, A-5020 Salzburg, Austria
| | - Hans Brandstetter
- Department of Molecular Biology and Christian Doppler Laboratory for Biosimilar Research, University of Salzburg, A-5020 Salzburg, Austria
| |
Collapse
|
38
|
Soh WT, Briza P, Dall E, Asam C, Schubert M, Huber S, Aglas L, Bohle B, Ferreira F, Brandstetter H. Two Distinct Conformations in Bet v 2 Determine Its Proteolytic Resistance to Cathepsin S. Int J Mol Sci 2017; 18:ijms18102156. [PMID: 29035299 PMCID: PMC5666837 DOI: 10.3390/ijms18102156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/04/2017] [Accepted: 10/11/2017] [Indexed: 12/17/2022] Open
Abstract
Birch pollen allergy affects more than 20% of the European allergic population. On a molecular level, birch pollen allergy can be linked to the two dominant allergens Bet v 1 and Bet v 2. Bet v 2 belongs to the profilin family, which is abundant in the plant kingdom. Importantly, the homologous plant profilins have a conserved cysteine motif with a currently unknown functional relevance. In particular, it is unknown whether the motif is relevant for disulfide formation and to what extent it would affect the profilins’ structural, functional and immunological properties. Here we present crystal structures of Bet v 2 in the reduced and the oxidized state, i.e., without and with a disulfide bridge. Despite overall structural similarity, the two structures distinctly differ at their termini which are stabilized to each other in the oxidized, i.e., disulfide-linked state. These structural differences translate into differences in their proteolytic resistance. Whereas the oxidized Bet v 2 is rather resistant towards the endolysosomal protease cathepsin S, it is rapidly degraded in the reduced form. By contrast, both Bet v 2 forms exhibit similar immunological properties as evidenced by their binding to IgE antibodies from birch pollen allergic patients and by their ability to trigger histamine release in a humanized rat basophilic leukemia cells (RBL) assay, independent of the presence or absence of the disulfide bridge. Taken together our findings suggest that the oxidized Bet v 2 conformation should be the relevant species, with a much longer retention time to trigger immune responses.
Collapse
Affiliation(s)
- Wai Tuck Soh
- Department of Molecular Biology, University of Salzburg, Salzburg 5020, Austria.
| | - Peter Briza
- Department of Molecular Biology, University of Salzburg, Salzburg 5020, Austria.
| | - Elfriede Dall
- Department of Molecular Biology, University of Salzburg, Salzburg 5020, Austria.
| | - Claudia Asam
- Department of Molecular Biology, University of Salzburg, Salzburg 5020, Austria.
| | - Mario Schubert
- Department of Molecular Biology, University of Salzburg, Salzburg 5020, Austria.
| | - Sara Huber
- Department of Molecular Biology, University of Salzburg, Salzburg 5020, Austria.
| | - Lorenz Aglas
- Department of Molecular Biology, University of Salzburg, Salzburg 5020, Austria.
| | - Barbara Bohle
- Department of Pathophysiology, Medical University of Vienna, Vienna 1090, Austria.
| | - Fatima Ferreira
- Department of Molecular Biology, University of Salzburg, Salzburg 5020, Austria.
| | - Hans Brandstetter
- Department of Molecular Biology, University of Salzburg, Salzburg 5020, Austria.
| |
Collapse
|
39
|
Elsässer B, Zauner FB, Messner J, Soh WT, Dall E, Brandstetter H. Distinct Roles of Catalytic Cysteine and Histidine in the Protease and Ligase Mechanisms of Human Legumain As Revealed by DFT-Based QM/MM Simulations. ACS Catal 2017; 7:5585-5593. [PMID: 28932620 PMCID: PMC5600538 DOI: 10.1021/acscatal.7b01505] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/10/2017] [Indexed: 11/30/2022]
Abstract
![]()
The cysteine protease enzyme legumain hydrolyzes peptide bonds
with high specificity after asparagine and under more acidic conditions
after aspartic acid [BakerE. N.J. Mol. Biol.1980, 141, 441−4847003158; BakerE. N.; J. Mol. Biol.1977, 111, 207–210859183; DrenthJ.; Biochemistry1976, 15, 3731–3738952885; MenardR.; J. Cell. Biochem.1994, 137; PolgarL.Eur. J. Biochem.1978, 88, 513–521689035; StorerA. C.; Methods Enzymol.1994, 244, 486–5007845227. Remarkably,
legumain additionally exhibits ligase activity that prevails at pH
> 5.5. The atomic reaction mechanisms including their pH dependence
are only partly understood. Here we present a density functional theory
(DFT)-based quantum mechanics/molecular mechanics (QM/MM) study of
the detailed reaction mechanism of both activities for human legumain
in solution. Contrasting the situation in other papain-like proteases,
our calculations reveal that the active site Cys189 must be present
in the protonated state for a productive nucleophilic attack and simultaneous
rupture of the scissile peptide bond, consistent with the experimental
pH profile of legumain-catalyzed cleavages. The resulting thioester
intermediate (INT1) is converted by water attack on the thioester
into a second intermediate, a diol (INT2), which is released by proton
abstraction by Cys189. Surprisingly, we found that ligation is not
the exact reverse of the proteolysis but can proceed via two distinct
routes. Whereas the transpeptidation route involves aminolysis of
the thioester (INT1), at pH 6 a cysteine-independent, histidine-assisted
ligation route was found. Given legumain’s important roles
in immunity, cancer, and neurodegenerative diseases, our findings
open up possibilities for targeted drug design in these fields.
Collapse
Affiliation(s)
- Brigitta Elsässer
- Department
of Molecular Biology, University of Salzburg, Billrothstrasse 11, A-5020 Salzburg, Austria
| | - Florian B. Zauner
- Department
of Molecular Biology, University of Salzburg, Billrothstrasse 11, A-5020 Salzburg, Austria
| | - Johann Messner
- Information
Management, University of Linz, Alternberger Strasse 69, A-4040 Linz, Austria
| | - Wai Tuck Soh
- Department
of Molecular Biology, University of Salzburg, Billrothstrasse 11, A-5020 Salzburg, Austria
| | - Elfriede Dall
- Department
of Molecular Biology, University of Salzburg, Billrothstrasse 11, A-5020 Salzburg, Austria
| | - Hans Brandstetter
- Department
of Molecular Biology, University of Salzburg, Billrothstrasse 11, A-5020 Salzburg, Austria
| |
Collapse
|
40
|
Lunde NN, Haugen MH, Bodin Larsen KB, Damgaard I, Pettersen SJ, Kasem R, Rut W, Drag M, Poreba M, Johansen HT, Solberg R. Glycosylation is important for legumain localization and processing to active forms but not for cystatin E/M inhibitory functions. Biochimie 2017; 139:27-37. [DOI: 10.1016/j.biochi.2017.05.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 05/15/2017] [Indexed: 12/28/2022]
|
41
|
Vidmar R, Vizovišek M, Turk D, Turk B, Fonović M. Protease cleavage site fingerprinting by label-free in-gel degradomics reveals pH-dependent specificity switch of legumain. EMBO J 2017; 36:2455-2465. [PMID: 28733325 DOI: 10.15252/embj.201796750] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 06/29/2017] [Accepted: 07/03/2017] [Indexed: 01/09/2023] Open
Abstract
Determination of protease specificity is of crucial importance for understanding protease function. We have developed the first gel-based label-free proteomic approach (DIPPS-direct in-gel profiling of protease specificity) that enables quick and reliable determination of protease cleavage specificities under large variety of experimental conditions. The methodology is based on in-gel digestion of the gel-separated proteome with the studied protease, enrichment of cleaved peptides by gel extraction, and subsequent mass spectrometry analysis combined with a length-limited unspecific database search. We applied the methodology to profile ten proteases ranging from highly specific (trypsin, endoproteinase GluC, caspase-7, and legumain) to broadly specific (matrix-metalloproteinase-3, thermolysin, and cathepsins K, L, S, and V). Using DIPPS, we were able to perform specificity profiling of thermolysin at its optimal temperature of 75°C, which confirmed the applicability of the method to extreme experimental conditions. Moreover, DIPPS enabled the first global specificity profiling of legumain at pH as low as 4.0, which revealed a pH-dependent change in the specificity of this protease, further supporting its broad applicability.
Collapse
Affiliation(s)
- Robert Vidmar
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia.,International Postgraduate School Jožef Stefan, Ljubljana, Slovenia
| | - Matej Vizovišek
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Dušan Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia.,International Postgraduate School Jožef Stefan, Ljubljana, Slovenia.,Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, Ljubljana, Slovenia
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia .,Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, Ljubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Marko Fonović
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia .,Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, Ljubljana, Slovenia
| |
Collapse
|
42
|
Reséndiz-Cardiel G, Arroyo R, Ortega-López J. Expression of the enzymatically active legumain-like cysteine proteinase TvLEGU-1 of Trichomonas vaginalis in Pichia pastoris. Protein Expr Purif 2017; 134:104-113. [DOI: 10.1016/j.pep.2017.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/04/2017] [Accepted: 04/14/2017] [Indexed: 01/11/2023]
|
43
|
Zhang Z, Obianyo O, Dall E, Du Y, Fu H, Liu X, Kang SS, Song M, Yu SP, Cabrele C, Schubert M, Li X, Wang JZ, Brandstetter H, Ye K. Inhibition of delta-secretase improves cognitive functions in mouse models of Alzheimer's disease. Nat Commun 2017; 8:14740. [PMID: 28345579 PMCID: PMC5378956 DOI: 10.1038/ncomms14740] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 01/26/2017] [Indexed: 12/21/2022] Open
Abstract
δ-secretase, also known as asparagine endopeptidase (AEP) or legumain, is a lysosomal cysteine protease that cleaves both amyloid precursor protein (APP) and tau, mediating the amyloid-β and tau pathology in Alzheimer's disease (AD). Here we report the therapeutic effect of an orally bioactive and brain permeable δ-secretase inhibitor in mouse models of AD. We performed a high-throughput screen and identified a non-toxic and selective δ-secretase inhibitor, termed compound 11, that specifically blocks δ-secretase but not other related cysteine proteases. Co-crystal structure analysis revealed a dual active site-directed and allosteric inhibition mode of this compound class. Chronic treatment of tau P301S and 5XFAD transgenic mice with this inhibitor reduces tau and APP cleavage, ameliorates synapse loss and augments long-term potentiation, resulting in protection of memory. Therefore, these findings demonstrate that this δ-secretase inhibitor may be an effective clinical therapeutic agent towards AD.
Collapse
Affiliation(s)
- Zhentao Zhang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA.,Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Obiamaka Obianyo
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Elfriede Dall
- Department of Molecular Biology, University of Salzburg, Salzburg A-5020, Austria
| | - Yuhong Du
- Department of Pharmacology, Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Haian Fu
- Department of Pharmacology, Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Seong Su Kang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Mingke Song
- Department of Anesthesiology Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Shan-Ping Yu
- Department of Anesthesiology Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Chiara Cabrele
- Department of Molecular Biology, University of Salzburg, Salzburg A-5020, Austria
| | - Mario Schubert
- Department of Molecular Biology, University of Salzburg, Salzburg A-5020, Austria
| | - Xiaoguang Li
- Pathophysiology Department, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian-Zhi Wang
- Pathophysiology Department, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Co-innovation Center of Neuroregeneration, Nantong 226001, China
| | - Hans Brandstetter
- Department of Molecular Biology, University of Salzburg, Salzburg A-5020, Austria
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
44
|
Yamane T, Kozuka M, Yamamoto Y, Nakano Y, Nakagaki T, Ohkubo I, Ariga H. Protease activity of legumain is inhibited by an increase of cystatin E/M in the DJ-1-knockout mouse spleen, cerebrum and heart. Biochem Biophys Rep 2017; 9:187-192. [PMID: 28956004 PMCID: PMC5614579 DOI: 10.1016/j.bbrep.2016.12.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 11/28/2016] [Accepted: 12/19/2016] [Indexed: 11/15/2022] Open
Abstract
Legumain (EC 3.4.22.34) is an asparaginyl endopeptidase. Legumain activity has been detected in various mouse tissues including the kidney, spleen and epididymis. Legumain is overexpressed in the majority of human solid tumors and transcription of the legumain gene is regulated by the p53 tumor suppressor in HCT116 cells. The legumain activity is also increased under acid conditions in Alzheimer's disease brains. DJ-1/PARK7, a cancer- and Parkinson's disease-associated protein, works as a coactivator to various transcription factors, including the androgen receptor, p53, PSF, Nrf2, SREBP and RREB1. Recently, we found that legumain expression, activation and cleavage of annexin A2 are regulated by DJ-1 through p53. In this study, we found that the expression levels of legumain mRNA were increased in the cerebrum, kidney, spleen, heart, lung, epididymis, stomach, small intestine and pancreas from DJ-1-knockout mice, although legumain activity levels were decreased in the cerebrum, spleen and heart from DJ-1-knockout mice. Furthermore, we found that cystatin E/M expression was increased in the spleen, cerebrum and heart from DJ-1-knockout mice. These results suggest that reduction of legumain activity is caused by an increase of cystatin E/M expression in the spleen, cerebrum and heart from DJ-1-knockout mice. Legumain is strongly activated in the epididymis from DJ-1-knockout mice. Expression level of legumain mRNA is increased but activity is decreased in the spleen, cerebrum and heart from DJ-1-knockout mice. Expression level of cystatin E/M is increased in the spleen, cerebrum and heart from DJ-1-knockout mice.
Collapse
Affiliation(s)
- Takuya Yamane
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| | - Miyuki Kozuka
- Department of Health and Nutrition, Faculty of Human Science, Hokkaido Bunkyo University, Eniwa 061-1449, Japan
| | - Yoshio Yamamoto
- Laboratory of Environmental Chemistry, Mie University Iga Research Institute, Yumegaoka, Iga 518-0131, Japan
| | - Yoshihisa Nakano
- Center for Research and Development Bioresources, Research Organization for University-Community Collaborations, Osaka Prefecture University, Sakai, Osaka 599-8570, Japan
| | - Takenori Nakagaki
- Institute of Food Sciences, Nakagaki Consulting Engineer and Co., Ltd, Nishi-ku, Sakai 593-8328, Japan
| | - Iwao Ohkubo
- Department of Nutrition, School of Nursing and Nutrition, Tenshi College, Higashi-ku, Sapporo 065-0013, Japan
| | - Hiroyoshi Ariga
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| |
Collapse
|
45
|
Szewińska J, Simińska J, Bielawski W. The roles of cysteine proteases and phytocystatins in development and germination of cereal seeds. JOURNAL OF PLANT PHYSIOLOGY 2016; 207:10-21. [PMID: 27771502 DOI: 10.1016/j.jplph.2016.09.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Proteolysis is an important process for development and germination of cereal seeds. Among the many types of proteases identified in plants are the cysteine proteases (CPs) of the papain and legumain families, which play a crucial role in hydrolysing storage proteins during seed germination as well as in processing the precursors of these proteins and the inactive forms of other proteases. Moreover, all of the tissues of cereal seeds undergo progressive degradation via programed cell death, which is integral to their growth. In view of the important roles played by proteases, their uncontrolled activity could be harmful to the development of seeds and young seedlings. Thus, the activities of these enzymes are regulated by intracellular inhibitors called phytocystatins (PhyCys). The phytocystatins inhibit the activity of proteases of the papain family, and the presence of an additional motif in their C-termini allows them to also regulate the activity of members of the legumain family. A balance between the levels of cysteine proteases and phytocystatins is necessary for proper cereal seed development, and this is maintained through the antagonistic activities of gibberellins (GAs) and abscisic acid (ABA), which regulate the expression of the corresponding genes. Transcriptional regulation of cysteine proteases and phytocystatins is determined by cis-acting elements located in the promoters of these genes and by the expression of their corresponding transcription factors (TFs) and the interactions between different TFs.
Collapse
Affiliation(s)
- Joanna Szewińska
- Warsaw University of Life Sciences-SGGW, Faculty of Agriculture and Biology, Department of Biochemistry, Nowoursynowska 159 street, Warsaw 02-776, Poland.
| | - Joanna Simińska
- Warsaw University of Life Sciences-SGGW, Faculty of Agriculture and Biology, Department of Biochemistry, Nowoursynowska 159 street, Warsaw 02-776, Poland
| | - Wiesław Bielawski
- Warsaw University of Life Sciences-SGGW, Faculty of Agriculture and Biology, Department of Biochemistry, Nowoursynowska 159 street, Warsaw 02-776, Poland
| |
Collapse
|
46
|
Lunde NN, Holm S, Dahl TB, Elyouncha I, Sporsheim B, Gregersen I, Abbas A, Skjelland M, Espevik T, Solberg R, Johansen HT, Halvorsen B. Increased levels of legumain in plasma and plaques from patients with carotid atherosclerosis. Atherosclerosis 2016; 257:216-223. [PMID: 27940038 DOI: 10.1016/j.atherosclerosis.2016.11.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 11/14/2016] [Accepted: 11/23/2016] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND AIMS The cysteine protease legumain has been shown to be up-regulated in unstable atherosclerotic plaques. This study aims to further elucidate legumain in atherosclerosis, by examining legumain in plasma and carotid plaques from patients with carotid stenosis. Furthermore, legumain secretion from monocyte-derived macrophages treated with atherogenic lipids during macrophage polarization was studied. METHODS Plasma levels of legumain from patients with carotid stenosis (n = 254), healthy controls (n = 91), and secreted from monocyte-derived macrophages were assessed by enzyme-linked-immunosorbent assay. Quantitative PCR and immunoblotting of legumain were performed on isolated plaques and legumain localization was visualized by immunohistochemistry and fluorescence microscopy. Monocyte-derived macrophages polarized to M1 or M2 macrophages were treated with VLDL, oxLDL or cholesterol crystals (CC) and the level of legumain analysed. RESULTS Patients with carotid stenosis had significantly higher levels of plasma legumain compared with healthy controls (median 2.0 versus 1.5 ng/ml, respectively; p = 0.003), although there was no correlation between the level of legumain and the degree of stenosis, and legumain was not an independent factor to identify patients with carotid plaques. Moreover, patients with symptoms the last 2 months had higher expressions of mature legumain, cystatin C and E/M, and the macrophage markers CD80 (M1) and CD163 (M2). Legumain co-localized with both M1 and M2 macrophages within plaques, whereas legumain mRNA expression was significantly higher (p < 0.0001) in plaques compared to non-atherosclerotic arteries (controls). Furthermore, in vitro studies showed significantly increased secretion of legumain from pro-inflammatory M1 compared to pro-resolving M2 macrophages (p = 0.014), and particularly in M1 treated with CC. In plaques, legumain was localized to structures resembling foam cells. CONCLUSIONS Legumain is increased in both plasma and plaques of patients with carotid stenosis and might be a new and early biomarker of atherosclerosis.
Collapse
Affiliation(s)
- Ngoc Nguyen Lunde
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Sverre Holm
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Hospital for Rheumatic Diseases, Lillehammer, Norway
| | - Tuva B Dahl
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| | - Inass Elyouncha
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Bjørnar Sporsheim
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Azhar Abbas
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Department of Neurology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Østfold Hospital Trust, Kalnes, Norway
| | - Mona Skjelland
- Department of Neurology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Terje Espevik
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rigmor Solberg
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway.
| | | | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| |
Collapse
|
47
|
Edgington-Mitchell LE, Wartmann T, Fleming AK, Gocheva V, van der Linden WA, Withana NP, Verdoes M, Aurelio L, Edgington-Mitchell D, Lieu T, Parker BS, Graham B, Reinheckel T, Furness JB, Joyce JA, Storz P, Halangk W, Bogyo M, Bunnett NW. Legumain is activated in macrophages during pancreatitis. Am J Physiol Gastrointest Liver Physiol 2016; 311:G548-60. [PMID: 27514475 PMCID: PMC5075999 DOI: 10.1152/ajpgi.00047.2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/18/2016] [Indexed: 01/31/2023]
Abstract
Pancreatitis is an inflammatory disease of the pancreas characterized by dysregulated activity of digestive enzymes, necrosis, immune infiltration, and pain. Repeated incidence of pancreatitis is an important risk factor for pancreatic cancer. Legumain, a lysosomal cysteine protease, has been linked to inflammatory diseases such as atherosclerosis, stroke, and cancer. Until now, legumain activation has not been studied during pancreatitis. We used a fluorescently quenched activity-based probe to assess legumain activation during caerulein-induced pancreatitis in mice. We detected activated legumain by ex vivo imaging, confocal microscopy, and gel electrophoresis. Compared with healthy controls, legumain activity in the pancreas of caerulein-treated mice was increased in a time-dependent manner. Legumain was localized to CD68(+) macrophages and was not active in pancreatic acinar cells. Using a small-molecule inhibitor of legumain, we found that this protease is not essential for the initiation of pancreatitis. However, it may serve as a biomarker of disease, since patients with chronic pancreatitis show strongly increased legumain expression in macrophages. Moreover, the occurrence of legumain-expressing macrophages in regions of acinar-to-ductal metaplasia suggests that this protease may influence reprogramming events that lead to inflammation-induced pancreatic cancer.
Collapse
Affiliation(s)
| | - Thomas Wartmann
- Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Alicia K Fleming
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, Florida
| | - Vasilena Gocheva
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Nimali P Withana
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud UMC, Nijmegen, The Netherlands
| | - Luigi Aurelio
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Daniel Edgington-Mitchell
- Laboratory for Turbulence Research in Aerospace and Combustion, Department of Mechanical and Aerospace Engineering, Monash University, Melbourne, Victoria, Australia
| | - TinaMarie Lieu
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Belinda S Parker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Bim Graham
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - John B Furness
- Department of Anatomy and Neuroscience, University of Melbourne and Florey Institute of Neuroscience and Mental Health, Parkville, Victoria Australia
| | - Johanna A Joyce
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Jacksonville, Florida
| | - Walter Halangk
- Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Nigel W Bunnett
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia; Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia; and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
48
|
Zhu W, Shao Y, Yang M, Jia M, Peng Y. Asparaginyl endopeptidase promotes proliferation and invasiveness of prostate cancer cells via PI3K/AKT signaling pathway. Gene 2016; 594:176-182. [PMID: 27590439 DOI: 10.1016/j.gene.2016.08.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 08/01/2016] [Accepted: 08/29/2016] [Indexed: 12/27/2022]
Abstract
Recurrence and metastasis are the major lethal causes of prostate cancer. It is urgent to find out the mechanisms and key factors governing prostate cancer progression and metastasis for developing new therapeutic strategies. Asparaginyl endopeptidase (AEP) overexpression has been found in a number of solid tumors. In prostate cancer, AEP has also been shown to exhibit a vesicular staining pattern and significantly associated with advanced tumor stage, high Gleason score, perineural invasion, and larger tumor. Here, we found that AEP was differentially expressed in prostate cancer cells with higher expression in 22RV1 cells and lower expression in PC-3 cells. AEP knockdown in 22RV1 cells significantly inhibited cell proliferation and invasion abilities while overexpression of AEP in PC-3 cells prompted cell proliferation and invasion abilities. Meanwhile, AEP knockdown upregulated cell apoptosis and vice versa. Further, we firstly identified that AEP promotes activation of the PI3K-AKT signaling pathway in prostate cancer cells. Taken together, our results suggest that AEP may be an attractive target for prostate cancer therapy.
Collapse
Affiliation(s)
- Wenjing Zhu
- Department of Urology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiqun Shao
- Department of Urology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ming Yang
- Department of Urology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Moran Jia
- Department of Urology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Peng
- Department of Urology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
49
|
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: 4.6] [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
|
50
|
Herrou J, Choi VM, Bubeck Wardenburg J, Crosson S. Activation Mechanism of the Bacteroides fragilis Cysteine Peptidase, Fragipain. Biochemistry 2016; 55:4077-84. [PMID: 27379832 DOI: 10.1021/acs.biochem.6b00546] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Enterotoxigenic Bacteroides fragilis produces a secreted metalloprotease known as B. fragilis toxin (BFT), which contributes to anaerobic sepsis, colitis, and colonic malignancy in mouse models of disease. A C11 family cysteine protease, fragipain (Fpn), directly activates BFT in the B. fragilis cell by removing the BFT prodomain. Fpn is itself a proenzyme and is autoactivated upon cleavage at an arginine residue in its activation loop. We have defined the proteolytic active site of Fpn, demonstrated that Fpn autoactivation can occur by an in trans loop cleavage mechanism, and characterized structural features of the Fpn activation loop that control peptidase activity against several substrates, including BFT. An arginine residue at the autocleavage site determines the fast activation kinetics of Fpn relative to the homologous C11 protease, PmC11, which is cleaved at lysine. Arginine to alanine substitution at the cleavage site ablated peptidase activity, as did partial truncation of the Fpn activation loop. However, complete truncation of the activation loop yielded an uncleaved, pro form of Fpn that was active as a peptidase against both Fpn and BFT substrates. Thus, Fpn can be transformed into an active peptidase in the absence of activation loop cleavage. This study provides insight into the mechanism of fragipain activation and, more generally, defines the role of the C11 activation loop in the control of peptidase activity and substrate specificity.
Collapse
Affiliation(s)
- Julien Herrou
- Department of Biochemistry and Molecular Biology, ‡Department of Microbiology, and §Department of Pediatrics, University of Chicago , 929 East 57th Street, Chicago, Illinois 60637, United States
| | - Vivian M Choi
- Department of Biochemistry and Molecular Biology, ‡Department of Microbiology, and §Department of Pediatrics, University of Chicago , 929 East 57th Street, Chicago, Illinois 60637, United States
| | - Juliane Bubeck Wardenburg
- Department of Biochemistry and Molecular Biology, ‡Department of Microbiology, and §Department of Pediatrics, University of Chicago , 929 East 57th Street, Chicago, Illinois 60637, United States
| | - Sean Crosson
- Department of Biochemistry and Molecular Biology, ‡Department of Microbiology, and §Department of Pediatrics, University of Chicago , 929 East 57th Street, Chicago, Illinois 60637, United States
| |
Collapse
|