1
|
Zhi M, Xie Y, Li H, Deng Z, Peng H, Yu Y, Zhang B. TGase-induced crosslinking of mulberry leaf protein particles as stabilizer of high-internal-phase Pickering emulsions: characterization and stability. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:4388-4399. [PMID: 40022672 DOI: 10.1002/jsfa.14196] [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: 11/11/2024] [Revised: 01/07/2025] [Accepted: 01/25/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Mulberry leaf protein (MLP) is a high-quality protein with significant nutritional value and functional properties. Enzymatic modification of proteins can enhance their functional properties by using proteases to covalently crosslink or hydrolyze proteins. This study investigates the potential of transglutaminase (TGase)-induced crosslinked MLP as an emulsifier in the formation of high-internal-phase Pickering emulsions. RESULTS Crosslinked MLP samples were prepared with TGase concentrations ranging from 0 to 25 U g-1. High-internal-phase Pickering emulsions (80% v/v) were formed at pH 8, with a crosslinking temperature of 50 °C, a TGase concentration of 20 U g-1 and an optimal crosslinking time of 60 min. As the enzyme concentration increased, the content of exposed sulfhydryl groups progressively increased, while the total free sulfhydryl content remained relatively stable. After varying crosslinking durations, both total free and exposed sulfhydryl group contents initially increased before declining. Additionally, the content of free amino groups in MLP gradually decreased with higher enzyme dosages and longer crosslinking times. The surface hydrophobicity of crosslinked MLP increased initially, followed by a decrease, reflecting changes in the spatial structure of MLP. SDS-PAGE analysis confirmed the formation of polymer masses after TGase-catalyzed crosslinking. Under optimal crosslinking conditions, the high-internal-phase Pickering emulsion prepared with TGase-induced crosslinked MLP exhibited a relatively uniform droplet distribution. CONCLUSION TGase-induced crosslinking enhances both the emulsifying activity and stability of MLP emulsions. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Mingze Zhi
- State Key Laboratory of Food Science and Resource, Nanchang University, Nanchang, China
| | - Yingshan Xie
- State Key Laboratory of Food Science and Resource, Nanchang University, Nanchang, China
| | - Hongyan Li
- State Key Laboratory of Food Science and Resource, Nanchang University, Nanchang, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resource, Nanchang University, Nanchang, China
| | - Han Peng
- Department of Food Science and Technology, University of California, Davis, CA, USA
| | - Yanfang Yu
- Jiangxi Cash Crops Institute, Nanchang, China
| | - Bing Zhang
- State Key Laboratory of Food Science and Resource, Nanchang University, Nanchang, China
| |
Collapse
|
2
|
Ryan B, Rangaswamy AMM, Shad S, Keillor JW. Diamino variants of piperazine-based tissue transglutaminase inhibitors. Bioorg Med Chem Lett 2025; 119:130078. [PMID: 39708924 DOI: 10.1016/j.bmcl.2024.130078] [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: 10/07/2024] [Revised: 11/29/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Tissue transglutaminase (TG2) is a multifunctional protein that can catalyze the cross-linking between proteins, and function as a G-protein. TG2's unregulated behaviour has been associated with fibrosis, celiac disease and cancer metastasis. Recently, small molecule irreversible inhibitors have been designed, bearing an electrophilic warhead that can react with the catalytic cysteine, abolishing TG2's catalytic and G-protein capabilities. Several research groups have converged on inhibitors comprising piperazine scaffolds, but no structure-activity relationships (SAR) of the piperazine core have been reported. In this study we synthesize a series of inhibitors with various diamino linkers, to understand what structural requirements are necessary for the core to help align the terminal acrylamide warhead in the optimal position. Kinetic evaluation using an in vitro biochemical assay provided the kinetic parameters kinact and KI for each inhibitor. This study revealed that adding a methyl group to the piperazine core can improve inhibitor efficiency.
Collapse
Affiliation(s)
- Brianna Ryan
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Alana M M Rangaswamy
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Sammir Shad
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada.
| |
Collapse
|
3
|
Iversen R, Heggelund JE, Das S, Høydahl LS, Sollid LM. Enzyme-activating B-cell receptors boost antigen presentation to pathogenic T cells in gluten-sensitive autoimmunity. Nat Commun 2025; 16:2387. [PMID: 40064932 PMCID: PMC11894174 DOI: 10.1038/s41467-025-57564-5] [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: 09/23/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Autoantibodies against the enzyme transglutaminase 3 (TG3) are characteristic to the gluten-sensitive skin disorder dermatitis herpetiformis (DH), which is an extraintestinal manifestation of celiac disease. We here demonstrate that TG3-specific B cells can activate gluten-specific CD4+ T cells through B-cell receptor (BCR)-mediated internalization of TG3-gluten enzyme-substrate complexes. Stereotypic anti-TG3 antibodies using IGHV2-5/IGKV4-1 gene segments enhance the catalytic activity of TG3, and this effect translates into increased gluten presentation to T cells when such antibodies are expressed as BCRs. The crystal structure of TG3 bound to an IGHV2-5/IGKV4-1 Fab shows that antibody binding to a β-sheet in the catalytic core domain causes the enzyme to adopt the active conformation. This mechanism explains the production of stereotypic anti-TG3 autoantibodies in DH and highlights a role for TG3-specific B cells as antigen-presenting cells for gluten-specific T cells. Similar boosting effects of autoreactive BCRs could be relevant for other autoimmune diseases, including rheumatoid arthritis.
Collapse
Affiliation(s)
- Rasmus Iversen
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| | - Julie Elisabeth Heggelund
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Saykat Das
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Lene S Høydahl
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
- Nextera AS, Oslo, Norway
| | - Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| |
Collapse
|
4
|
Kang J, Jeon HY, Lee J, Bae S, Park GY, Min KJ, Joo J, Lee AJ, Kim HJ, Im CY, Kim EB, Lee JH, Hwang JS, Lee S, Lee JY, Navals P, Keillor JW, Ha KS, Song M. Structurally Minimalized and Druglike TGase2 Inhibitors Based on 7-Aminoquinoline-5,8-dione Scaffolds for the Treatment of Diabetic Retinopathy. J Med Chem 2024; 67:19716-19735. [PMID: 39445793 DOI: 10.1021/acs.jmedchem.4c02081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Diabetic retinopathy is a disease that can cause vision loss leading to blindness in people with diabetes. Improved methods to treat and prevent vision loss in diabetic patients are in high demand owing to limited current treatment procedures. Herein, we report a new class of transglutaminase 2 (TGase2) inhibitors for the treatment of diabetic retinopathy based on 7-aminoquinoline-5,8-dione derivatives. 7-Amino-2-phenylquinoline-5,8-dione 11 and 7-amino-2-{4-[(1-methylpiperidin-4-yl)oxy]phenyl}quinoline-5,8-dione 23 exhibited potent inhibitory activities against TGase2 in a fibrinogen array-based on-chip TGase2 activity assay and in an in situ assay in human retinal microvascular endothelial cells, with IC50 values of 5.88 and 1.12 μM in vitro, and 0.09 and 0.07 μM in situ, respectively. Pharmacokinetically favorable 7-amino-2-{4-[(1-isopropylpiperidin-4-yl)oxy] phenyl}quinoline-5,8-dione 22 inhibited vascular leakage in the retinas of streptozotocin-induced diabetic mice via oral administration. Results from the AL5 kinetic assay and a molecular docking study suggest that the inhibitors may bind to TGase2 remote from the active site.
Collapse
Affiliation(s)
- Jihee Kang
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Hye-Yoon Jeon
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Korea
- Scripps Korea Antibody Institute, Kangwon National University Chuncheon Campus, Chuncheon, Kangwon-do 24341, Korea
| | - Jieon Lee
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Seri Bae
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Ga Young Park
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Kyoung-Jin Min
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Jeongmin Joo
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Ah-Jun Lee
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Korea
| | - Hyo-Ji Kim
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Chun Young Im
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Eun-Bin Kim
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Korea
| | - Ji Hun Lee
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Ji Sun Hwang
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Seungju Lee
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Jee-Young Lee
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| | - Pauline Navals
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Korea
| | - Minsoo Song
- New Drug Discovery Center (NDDC), Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro Dong-gu, Daegu 41061, Korea
| |
Collapse
|
5
|
Rangaswamy AMM, Roy FM, Keillor JW. Small molecule substrates for the rapid quantification of acyl transfer activity of nylon hydrolase NylC A. Anal Biochem 2024; 693:115598. [PMID: 38964700 DOI: 10.1016/j.ab.2024.115598] [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: 05/06/2024] [Revised: 06/06/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
The widespread use of polyamides such as nylons has led to the accumulation of nylon waste, which is particularly resistant to decomposition due to the intrinsic stability of the amide bond. New methods are required for the true recycling of these waste materials by depolymerization. Enzymes that are capable of hydrolyzing polyamides have been proposed as biocatalysts that may be suitable for this application. NylC is an enzyme that can mediate the hydrolysis of aminohexanoic acid oligomers, and to some extent, bulk polymers. However, current assays to characterize the activity of this enzyme require long reaction times and/or rely on secondary reactions to quantify hydrolysis. Herein, we have designed structurally-optimized small molecule chromogenic esters that serve as substrate analogues for monitoring NylC acyltransferase activity in a continuous manner. This assay can be performed in minutes at room temperature, and the substrate N-acetyl-GABA-pNP ester (kcat = 0.37 s-1, KM = 256 μM) shows selectivity for NylC in complex biological media. We also demonstrate that activity towards this substrate analogue correlates with amide hydrolysis, which is the primary activity of this enzyme. Furthermore, our screening of substrate analogues provides insight into the substrate specificity of NylC, which is relevant to biocatalytic applications.
Collapse
Affiliation(s)
- Alana M M Rangaswamy
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Francis M Roy
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada.
| |
Collapse
|
6
|
Li H, Wu J, Zhang N, Zheng Q. Transglutaminase 2-mediated histone monoaminylation and its role in cancer. Biosci Rep 2024; 44:BSR20240493. [PMID: 39115570 PMCID: PMC11345673 DOI: 10.1042/bsr20240493] [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: 06/27/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/10/2024] Open
Abstract
Transglutaminase 2 (TGM2) has been known as a well-characterized factor regulating the progression of multiple types of cancer, due to its multifunctional activities and the ubiquitous signaling pathways it is involved in. As a member of the transglutaminase family, TGM2 catalyzes protein post-translational modifications (PTMs), including monoaminylation, amide hydrolysis, cross-linking, etc., through the transamidation of variant glutamine-containing protein substrates. Recent discoveries revealed histone as an important category of TGM2 substrates, thus identifying histone monoaminylation as an emerging epigenetic mark, which is highly enriched in cancer cells and possesses significant regulatory functions of gene transcription. In this review, we will summarize recent advances in TGM2-mediated histone monoaminylation as well as its role in cancer and discuss the key research methodologies to better understand this unique epigenetic mark, thereby shedding light on the therapeutic potential of TGM2 as a druggable target in cancer treatment.
Collapse
Affiliation(s)
- Huapeng Li
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH 43210, U.S.A
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, U.S.A
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, U.S.A
| | - Jinghua Wu
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, U.S.A
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, U.S.A
| | - Nan Zhang
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, U.S.A
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, U.S.A
| | - Qingfei Zheng
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH 43210, U.S.A
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, U.S.A
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, U.S.A
- Department of Biological Chemistry and Pharmacology, College of Medicine, The Ohio State University, Columbus, OH 43210, U.S.A
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, IN, U.S.A
| |
Collapse
|
7
|
Sewa AS, Besser HA, Mathews II, Khosla C. Structural and mechanistic analysis of Ca 2+-dependent regulation of transglutaminase 2 activity using a Ca 2+-bound intermediate state. Proc Natl Acad Sci U S A 2024; 121:e2407066121. [PMID: 38959038 PMCID: PMC11252922 DOI: 10.1073/pnas.2407066121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
Mammalian transglutaminases, a family of Ca2+-dependent proteins, are implicated in a variety of diseases. For example, celiac disease (CeD) is an autoimmune disorder whose pathogenesis requires transglutaminase 2 (TG2) to deamidate select glutamine residues in diet-derived gluten peptides. Deamidation involves the formation of transient γ-glutamyl thioester intermediates. Recent studies have revealed that in addition to the deamidated gluten peptides themselves, their corresponding thioester intermediates are also pathogenically relevant. A mechanistic understanding of this relevance is hindered by the absence of any structure of Ca2+-bound TG2. We report the X-ray crystallographic structure of human TG2 bound to an inhibitory gluten peptidomimetic and two Ca2+ ions in sites previously designated as S1 and S3. Together with additional structure-guided experiments, this structure provides a mechanistic explanation for how S1 regulates formation of an inhibitory disulfide bond in TG2, while also establishing that S3 is essential for γ-glutamyl thioester formation. Furthermore, our crystallographic findings and associated analyses have revealed that i) two interacting residues, H305 and E363, play a critical role in resolving the thioester intermediate into an isopeptide bond (transamidation) but not in thioester hydrolysis (deamidation); and ii) residues N333 and K176 stabilize preferred TG2 substrates and inhibitors via hydrogen bonding to nonreactive backbone atoms. Overall, the intermediate-state conformer of TG2 reported here represents a superior model to previously characterized conformers for both transition states of the TG2-catalyzed reaction.
Collapse
Affiliation(s)
- Agnele S. Sewa
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
| | - Harrison A. Besser
- Department of Chemistry, Stanford University, Stanford, CA94305
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA94305
| | - Irimpan I. Mathews
- Stanford Synchrotron Radiation Lightsource, Stanford Linear Accelerator Center National Accelerator Laboratory, Menlo Park, CA94025
| | - Chaitan Khosla
- Department of Chemistry, Stanford University, Stanford, CA94305
- Department of Chemical Engineering, Stanford University, Stanford, CA94305
- Sarafan Chemistry, Engineering Medicine and Human Health, Stanford University, Stanford, CA94305
| |
Collapse
|
8
|
Park HW, Lee CE, Kim S, Jeong WJ, Kim K. Ex Vivo Peptide Decoration Strategies on Stem Cell Surfaces for Augmenting Endothelium Interaction. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:327-339. [PMID: 37830185 DOI: 10.1089/ten.teb.2023.0210] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Ischemic vascular diseases remain leading causes of disability and death. Although various clinical therapies have been tried, reperfusion injury is a major issue, occurring when blood recirculates at the damaged lesion. As an alternative approach, cell-based therapy has emerged. Mesenchymal stem cells (MSCs) are attractive cellular candidates due to their therapeutic capacities, including differentiation, safety, angiogenesis, and tissue repair. However, low levels of receptors/ligands limit targeted migration of stem cells. Thus, it is important to improve homing efficacy of transplanted MSCs toward damaged endothelium. Among various MSC modulations, ex vivo cell surface engineering could effectively augment homing efficiency by decorating MSC surfaces with alternative receptors/ligands, thereby facilitating intercellular interactions with the endothelium. Especially, exogenous decoration of peptides onto stem cell surfaces could provide appropriate functional signaling moieties to achieve sufficient MSC homing. Based on their protein-like functionalities, high modularity in molecular design, and high specific affinities and multivalency to target receptors, peptides could be representative surface-presentable moieties. Moreover, peptides feature a mild synthetic process, enabling precise control of amino acid composition and sequence. Such ex vivo stem cell surface engineering could be achieved primarily by hydrophobic interactions of the cellular bilayer with peptide-conjugated anchor modules and by covalent conjugation between peptides and available compartments in membranes. To this end, this review provides an overview of currently available peptide-mediated, ex vivo stem cell surface engineering strategies for enhancing MSC homing efficiency by facilitating interactions with endothelial cells. Stem cell surface engineering techniques using peptide-based bioconjugates have the potential to revolutionize current vascular disease treatments while addressing their technical limitations.
Collapse
Affiliation(s)
- Hee Won Park
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul, Republic of Korea
| | - Chae Eun Lee
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul, Republic of Korea
| | - Sungjun Kim
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul, Republic of Korea
| | - Woo-Jin Jeong
- Department of Biological Engineering, Inha University, Incheon, Republic of Korea
| | - Kyobum Kim
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul, Republic of Korea
| |
Collapse
|
9
|
Navals P, Rangaswamy AMM, Kasyanchyk P, Berezovski MV, Keillor JW. Conformational Modulation of Tissue Transglutaminase via Active Site Thiol Alkylating Agents: Size Does Not Matter. Biomolecules 2024; 14:496. [PMID: 38672511 PMCID: PMC11048362 DOI: 10.3390/biom14040496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/02/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
TG2 is a unique member of the transglutaminase family as it undergoes a dramatic conformational change, allowing its mutually exclusive function as either a cross-linking enzyme or a G-protein. The enzyme's dysregulated activity has been implicated in a variety of pathologies (e.g., celiac disease, fibrosis, cancer), leading to the development of a wide range of inhibitors. Our group has primarily focused on the development of peptidomimetic targeted covalent inhibitors, the nature and size of which were thought to be important features to abolish TG2's conformational dynamism and ultimately inhibit both its activities. However, we recently demonstrated that the enzyme was unable to bind guanosine triphosphate (GTP) when catalytically inactivated by small molecule inhibitors. In this study, we designed a library of models targeting covalent inhibitors of progressively smaller sizes (15 to 4 atoms in length). We evaluated their ability to inactivate TG2 by measuring their respective kinetic parameters kinact and KI. Their impact on the enzyme's ability to bind GTP was then evaluated and subsequently correlated to the conformational state of the enzyme, as determined via native PAGE and capillary electrophoresis. All irreversible inhibitors evaluated herein locked TG2 in its open conformation and precluded GTP binding. Therefore, we conclude that steric bulk and structural complexity are not necessary factors to consider when designing TG2 inhibitors to abolish G-protein activity.
Collapse
Affiliation(s)
| | | | | | | | - Jeffrey W. Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (P.N.); (A.M.M.R.); (P.K.); (M.V.B.)
| |
Collapse
|
10
|
Buccarelli M, Castellani G, Fiorentino V, Pizzimenti C, Beninati S, Ricci-Vitiani L, Scattoni ML, Mischiati C, Facchiano F, Tabolacci C. Biological Implications and Functional Significance of Transglutaminase Type 2 in Nervous System Tumors. Cells 2024; 13:667. [PMID: 38667282 PMCID: PMC11048792 DOI: 10.3390/cells13080667] [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: 01/31/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Transglutaminase type 2 (TG2) is the most ubiquitously expressed member of the transglutaminase family. TG2 catalyzes the transamidation reaction leading to several protein post-translational modifications and it is also implicated in signal transduction thanks to its GTP binding/hydrolyzing activity. In the nervous system, TG2 regulates multiple physiological processes, such as development, neuronal cell death and differentiation, and synaptic plasticity. Given its different enzymatic activities, aberrant expression or activity of TG2 can contribute to tumorigenesis, including in peripheral and central nervous system tumors. Indeed, TG2 dysregulation has been reported in meningiomas, medulloblastomas, neuroblastomas, glioblastomas, and other adult-type diffuse gliomas. The aim of this review is to provide an overview of the biological and functional relevance of TG2 in the pathogenesis of nervous system tumors, highlighting its involvement in survival, tumor inflammation, differentiation, and in the resistance to standard therapies.
Collapse
Affiliation(s)
- Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Cristina Pizzimenti
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy;
| | - Simone Beninati
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Maria Luisa Scattoni
- Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Carlo Mischiati
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Claudio Tabolacci
- Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| |
Collapse
|
11
|
du Pre MF, Iversen R, Sollid LM. Coeliac disease: the paradox of diagnosing a food hypersensitivity disorder with autoantibodies. Gut 2024; 73:844-853. [PMID: 38378252 DOI: 10.1136/gutjnl-2023-331595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/30/2024] [Indexed: 02/22/2024]
Abstract
Serum antibodies to the autoantigen transglutaminase 2 (TG2) are increasingly harnessed to diagnose coeliac disease. Diagnostic guidelines for children give recommendation for a no-biopsy-based diagnosis through detection of high amounts of IgA anti-TG2 antibodies in serum with confirmation of positivity in a separate blood sample by characteristic autoantibody-staining of tissue. While measurement of IgA anti-TG2 also is important in the diagnostic workup of adults, the adult guidelines still mandate examination of gut biopsies. This requirement might well change in the future, as might the necessity for confirming autoantibody positivity by tissue staining. The key role of autoantibody serology for diagnosis of coeliac disease is paradoxical. Coeliac disease was considered, and still can be considered, a food intolerance disorder where autoantibodies at face value are out of place. The immunological mechanisms underlying the formation of autoantibodies in response to gluten exposure have been dissected. This review presents the current insights demonstrating that the autoantibodies in coeliac disease are intimately integrated in the maladapted immune response to gluten.
Collapse
Affiliation(s)
- M Fleur du Pre
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hosptial - Rikshospitalet, Oslo, Norway
| | - Rasmus Iversen
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hosptial - Rikshospitalet, Oslo, Norway
| | - Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hosptial - Rikshospitalet, Oslo, Norway
| |
Collapse
|
12
|
Liu J, Mouradian MM. Pathogenetic Contributions and Therapeutic Implications of Transglutaminase 2 in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:2364. [PMID: 38397040 PMCID: PMC10888553 DOI: 10.3390/ijms25042364] [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: 12/29/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Neurodegenerative diseases encompass a heterogeneous group of disorders that afflict millions of people worldwide. Characteristic protein aggregates are histopathological hallmark features of these disorders, including Amyloid β (Aβ)-containing plaques and tau-containing neurofibrillary tangles in Alzheimer's disease, α-Synuclein (α-Syn)-containing Lewy bodies and Lewy neurites in Parkinson's disease and dementia with Lewy bodies, and mutant huntingtin (mHTT) in nuclear inclusions in Huntington's disease. These various aggregates are found in specific brain regions that are impacted by neurodegeneration and associated with clinical manifestations. Transglutaminase (TG2) (also known as tissue transglutaminase) is the most ubiquitously expressed member of the transglutaminase family with protein crosslinking activity. To date, Aβ, tau, α-Syn, and mHTT have been determined to be substrates of TG2, leading to their aggregation and implicating the involvement of TG2 in several pathophysiological events in neurodegenerative disorders. In this review, we summarize the biochemistry and physiologic functions of TG2 and describe recent advances in the pathogenetic role of TG2 in these diseases. We also review TG2 inhibitors tested in clinical trials and discuss recent TG2-targeting approaches, which offer new perspectives for the design of future highly potent and selective drugs with improved brain delivery as a disease-modifying treatment for neurodegenerative disorders.
Collapse
Affiliation(s)
| | - M. Maral Mouradian
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
| |
Collapse
|
13
|
Wodtke R, Laube M, Hauser S, Meister S, Ludwig FA, Fischer S, Kopka K, Pietzsch J, Löser R. Preclinical evaluation of an 18F-labeled N ε-acryloyllysine piperazide for covalent targeting of transglutaminase 2. EJNMMI Radiopharm Chem 2024; 9:1. [PMID: 38165538 PMCID: PMC10761660 DOI: 10.1186/s41181-023-00231-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/15/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Transglutaminase 2 (TGase 2) is a multifunctional protein and has a prominent role in various (patho)physiological processes. In particular, its transamidase activity, which is rather latent under physiological conditions, gains importance in malignant cells. Thus, there is a great need of theranostic probes for targeting tumor-associated TGase 2, and targeted covalent inhibitors appear to be particularly attractive as vector molecules. Such an inhibitor, equipped with a radionuclide suitable for noninvasive imaging, would be supportive for answering the general question on the possibility for functional characterization of tumor-associated TGase 2. For this purpose, the recently developed 18F-labeled Nε-acryloyllysine piperazide [18F]7b, which is a potent and selective irreversible inhibitor of TGase 2, was subject to a detailed radiopharmacological characterization herein. RESULTS An alternative radiosynthesis of [18F]7b is presented, which demands less than 300 µg of the respective trimethylammonio precursor per synthesis and provides [18F]7b in good radiochemical yields (17 ± 7%) and high (radio)chemical purities (≥ 99%). Ex vivo biodistribution studies in healthy mice at 5 and 60 min p.i. revealed no permanent enrichment of 18F-activity in tissues with the exception of the bone tissue. In vivo pretreatment with ketoconazole and in vitro murine liver microsome studies complemented by mass spectrometric analysis demonstrated that bone uptake originates from metabolically released [18F]fluoride. Further metabolic transformations of [18F]7b include mono-hydroxylation and glucuronidation. Based on blood sampling data and liver microsome experiments, pharmacokinetic parameters such as plasma and intrinsic clearance were derived, which substantiated the apparently rapid distribution of [18F]7b in and elimination from the organisms. A TGase 2-mediated uptake of [18F]7b in different tumor cell lines could not be proven. Moreover, evaluation of [18F]7b in melanoma tumor xenograft models based on A375-hS100A4 (TGase 2 +) and MeWo (TGase 2 -) cells by ex vivo biodistribution and PET imaging studies were not indicative for a specific targeting. CONCLUSION [18F]7b is a valuable radiometric tool to study TGase 2 in vitro under various conditions. However, its suitability for targeting tumor-associated TGase 2 is strongly limited due its unfavorable pharmacokinetic properties as demonstrated in rodents. Consequently, from a radiochemical perspective [18F]7b requires appropriate structural modifications to overcome these limitations.
Collapse
Affiliation(s)
- Robert Wodtke
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany.
| | - Markus Laube
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Sandra Hauser
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Sebastian Meister
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Friedrich-Alexander Ludwig
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstraße 15, 04318, Leipzig, Germany
| | - Steffen Fischer
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstraße 15, 04318, Leipzig, Germany
| | - Klaus Kopka
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstraße 15, 04318, Leipzig, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069, Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069, Dresden, Germany
| | - Reik Löser
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany.
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069, Dresden, Germany.
| |
Collapse
|
14
|
Gates EWJ, Prince-Hallée A, Heidari Y, Sedighi A, Keillor JW. High-Affinity Fluorogenic Substrate for Tissue Transglutaminase Reveals Enzymatic Hysteresis. Biochemistry 2023; 62:3085-3095. [PMID: 37856791 DOI: 10.1021/acs.biochem.3c00337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Transglutaminases (TGases) are a family of calcium-dependent enzymes primarily known for their ability to cross-link proteins. Transglutaminase 2 (TG2) is one isozyme in this family whose role is multifaceted. TG2 can act not only as a typical transamidase through its catalytic core but also as a G-protein via its GTP binding site. These two discrete activities are tightly regulated by both environmental stimuli and redox reactions. Ubiquitously expressed in humans, TG2 has been implicated in numerous disease pathologies that require extensive investigation. The catalytic activity of TG2 can be monitored through various mechanisms, including hydrolysis, transamidation, or cleavage of isopeptide bonds. Activity assays are required to monitor the activity of this isozyme not only for studying its transamidation reaction but also for validation of therapeutics designed to abolish this activity. Herein, we present the design, synthesis, and evaluation of a new TG2 activity substrate based on a previously optimized inhibitor scaffold. The substrate APH7 exhibits excellent affinity, selectivity, and reactivity with TG2 (KM = 3.0 μM). Furthermore, its application also allowed the discovery of unique hysteresis at play within the catalytic activity and inhibition reactivity of TG2.
Collapse
Affiliation(s)
- Eric W J Gates
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Adrien Prince-Hallée
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Yasaman Heidari
- Dalriada Drug Discovery, Mississauga, Ontario L5N 8G4, Canada
| | | | - Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| |
Collapse
|
15
|
Heggelund JE, Das S, Stamnaes J, Iversen R, Sollid LM. Autoantibody binding and unique enzyme-substrate intermediate conformation of human transglutaminase 3. Nat Commun 2023; 14:6216. [PMID: 37798283 PMCID: PMC10556103 DOI: 10.1038/s41467-023-42004-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/25/2023] [Indexed: 10/07/2023] Open
Abstract
Transglutaminase 3 (TG3), the autoantigen of dermatitis herpetiformis (DH), is a calcium dependent enzyme that targets glutamine residues in polypeptides for either transamidation or deamidation modifications. To become catalytically active TG3 requires proteolytic cleavage between the core domain and two C-terminal β-barrels (C1C2). Here, we report four X-ray crystal structures representing inactive and active conformations of human TG3 in complex with a TG3-specific Fab fragment of a DH patient derived antibody. We demonstrate that cleaved TG3, upon binding of a substrate-mimicking inhibitor, undergoes a large conformational change as a β-sheet in the catalytic core domain moves and C1C2 detaches. The unique enzyme-substrate conformation of TG3 without C1C2 is recognized by DH autoantibodies. The findings support a model where B-cell receptors of TG3-specific B cells bind and internalize TG3-gluten enzyme-substrate complexes thereby facilitating gluten-antigen presentation, T-cell help and autoantibody production.
Collapse
Affiliation(s)
- Julie Elisabeth Heggelund
- KG Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway.
| | - Saykat Das
- KG Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Jorunn Stamnaes
- KG Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Rasmus Iversen
- KG Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Ludvig M Sollid
- KG Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway.
| |
Collapse
|
16
|
Gates EWJ, Calvert ND, Cundy NJ, Brugnoli F, Navals P, Kirby A, Bianchi N, Adhikary G, Shuhendler AJ, Eckert RL, Keillor JW. Cell-Impermeable Inhibitors Confirm That Intracellular Human Transglutaminase 2 Is Responsible for the Transglutaminase-Associated Cancer Phenotype. Int J Mol Sci 2023; 24:12546. [PMID: 37628729 PMCID: PMC10454375 DOI: 10.3390/ijms241612546] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
Transglutaminase 2 (TG2) is a multifunctional enzyme primarily responsible for crosslinking proteins. Ubiquitously expressed in humans, TG2 can act either as a transamidase by crosslinking two substrates through formation of an Nε(ɣ-glutaminyl)lysine bond or as an intracellular G-protein. These discrete roles are tightly regulated by both allosteric and environmental stimuli and are associated with dramatic changes in the conformation of the enzyme. The pleiotropic nature of TG2 and multi-faceted activities have resulted in TG2 being implicated in numerous disease pathologies including celiac disease, fibrosis, and cancer. Targeted TG2 therapies have not been selective for subcellular localization, such that currently no tools exist to selectively target extracellular over intracellular TG2. Herein, we have designed novel TG2-selective inhibitors that are not only highly potent and irreversible, but also cell impermeable, targeting only extracellular TG2. We have also further derivatized the scaffold to develop probes that are intrinsically fluorescent or bear an alkyne handle, which target both intra- and extracellular TG2, in order to facilitate cellular labelling and pull-down assays. The fluorescent probes were internalized and imaged in cellulo, and provide the first implicit experimental evidence that by comparison with their cell-impermeable analogues, it is specifically intracellular TG2, and presumably its G-protein activity, that contributes to transglutaminase-associated cancer progression.
Collapse
Affiliation(s)
- Eric W. J. Gates
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| | - Nicholas D. Calvert
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| | - Nicholas J. Cundy
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| | - Federica Brugnoli
- Department of Translational Medicine, University of Ferrara, 44021 Ferrara, Italy; (F.B.); (N.B.)
| | - Pauline Navals
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| | - Alexia Kirby
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, 44021 Ferrara, Italy; (F.B.); (N.B.)
| | - Gautam Adhikary
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.A.); (R.L.E.)
| | - Adam J. Shuhendler
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| | - Richard L. Eckert
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.A.); (R.L.E.)
| | - Jeffrey W. Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (E.W.J.G.); (N.D.C.); (N.J.C.); (P.N.); (A.K.); (A.J.S.)
| |
Collapse
|
17
|
Mader L, Watt SKI, Iyer HR, Nguyen L, Kaur H, Keillor JW. The war on hTG2: warhead optimization in small molecule human tissue transglutaminase inhibitors. RSC Med Chem 2023; 14:277-298. [PMID: 36846370 PMCID: PMC9945866 DOI: 10.1039/d2md00378c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022] Open
Abstract
Human tissue transglutaminase (hTG2) is a multifunctional enzyme with protein cross-linking and G-protein activity, both of which have been implicated in the progression of diseases such as fibrosis and cancer stem cell propagation when dysregulated, prompting the development of small molecule targeted covalent inhibitors (TCIs) possessing a crucial electrophilic 'warhead'. In recent years there have been significant advances in the library of warheads available for the design of TCIs; however, the exploration of warhead functionality in hTG2 inhibitors has remained relatively stagnant. Herein, we describe a structure-activity relationship study entailing rational design and synthesis for systematic variation of the warhead on a previously reported small molecule inhibitor scaffold, and rigorous kinetic evaluation of inhibitory efficiency, selectivity, and pharmacokinetic stability. This study reveals a strong influence on the kinetic parameters k inact and K I with even subtle variation in warhead structure, suggesting that the warhead plays a significant role in not only reactivity, but also binding affinity, which consequently extends to isozyme selectivity. Warhead structure also influences in vivo stability, which we model by measuring intrinsic reactivity with glutathione, as well as stability in hepatocytes and in whole blood, giving insight into degradation pathways and relative therapeutic potential of different functional groups. This work provides fundamental structural and reactivity information highlighting the importance of strategic warhead design for the development of potent hTG2 inhibitors.
Collapse
Affiliation(s)
- Lavleen Mader
- Department of Chemistry and Biomolecular Sciences, University of Ottawa Ottawa Ontario K1N 6N5 Canada
| | - Sarah K I Watt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa Ottawa Ontario K1N 6N5 Canada
| | - Harish R Iyer
- Dalriada Drug Discovery Mississauga Ontario L5N 8G4 Canada
| | - Linh Nguyen
- Dalriada Drug Discovery Mississauga Ontario L5N 8G4 Canada
| | - Harpreet Kaur
- Dalriada Drug Discovery Mississauga Ontario L5N 8G4 Canada
| | - Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa Ottawa Ontario K1N 6N5 Canada
| |
Collapse
|
18
|
Peptidic Inhibitors and a Fluorescent Probe for the Selective Inhibition and Labelling of Factor XIIIa Transglutaminase. Molecules 2023; 28:molecules28041634. [PMID: 36838622 PMCID: PMC9960274 DOI: 10.3390/molecules28041634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Factor XIIIa (FXIIIa) is a transglutaminase of major therapeutic interest for the development of anticoagulants due to its essential role in the blood coagulation cascade. While numerous FXIIIa inhibitors have been reported, they failed to reach clinical evaluation due to their lack of metabolic stability and low selectivity over transglutaminase 2 (TG2). Furthermore, the chemical tools available for the study of FXIIIa activity and localization are extremely limited. To combat these shortcomings, we designed, synthesised, and evaluated a library of 21 novel FXIIIa inhibitors. Electrophilic warheads, linker lengths, and hydrophobic units were varied on small molecule and peptidic scaffolds to optimize isozyme selectivity and potency. A previously reported FXIIIa inhibitor was then adapted for the design of a probe bearing a rhodamine B moiety, producing the innovative KM93 as the first known fluorescent probe designed to selectively label active FXIIIa with high efficiency (kinact/KI = 127,300 M-1 min-1) and 6.5-fold selectivity over TG2. The probe KM93 facilitated fluorescent microscopy studies within bone marrow macrophages, labelling FXIIIa with high efficiency and selectivity in cell culture. The structure-activity trends with these novel inhibitors and probes will help in the future study of the activity, inhibition, and localization of FXIIIa.
Collapse
|
19
|
The Impact of Nε-Acryloyllysine Piperazides on the Conformational Dynamics of Transglutaminase 2. Int J Mol Sci 2023; 24:ijms24021650. [PMID: 36675164 PMCID: PMC9865645 DOI: 10.3390/ijms24021650] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/17/2023] Open
Abstract
In addition to the classic functions of proteins, such as acting as a biocatalyst or binding partner, the conformational states of proteins and their remodeling upon stimulation need to be considered. A prominent example of a protein that undergoes comprehensive conformational remodeling is transglutaminase 2 (TGase 2), the distinct conformational states of which are closely related to particular functions. Its involvement in various pathophysiological processes, including fibrosis and cancer, motivates the development of theranostic agents, particularly based on inhibitors that are directed toward the transamidase activity. In this context, the ability of such inhibitors to control the conformational dynamics of TGase 2 emerges as an important parameter, and methods to assess this property are in great demand. Herein, we describe the application of the switchSENSE® principle to detect conformational changes caused by three irreversibly binding Nε-acryloyllysine piperazides, which are suitable radiotracer candidates of TGase 2. The switchSENSE® technique is based on DNA levers actuated by alternating electric fields. These levers are immobilized on gold electrodes with one end, and at the other end of the lever, the TGase 2 is covalently bound. A novel computational method is introduced for describing the resulting lever motion to quantify the extent of stimulated conformational TGase 2 changes. Moreover, as a complementary biophysical method, native polyacrylamide gel electrophoresis was performed under similar conditions to validate the results. Both methods prove the occurrence of an irreversible shift in the conformational equilibrium of TGase 2, caused by the binding of the three studied Nε-acryloyllysine piperazides.
Collapse
|
20
|
Müller CD, Ruiz-Gómez G, Cazzonelli S, Möller S, Wodtke R, Löser R, Freyse J, Dürig JN, Rademann J, Hempel U, Pisabarro MT, Vogel S. Sulfated glycosaminoglycans inhibit transglutaminase 2 by stabilizing its closed conformation. Sci Rep 2022; 12:13326. [PMID: 35922533 PMCID: PMC9349199 DOI: 10.1038/s41598-022-17113-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/20/2022] [Indexed: 11/09/2022] Open
Abstract
Transglutaminases (TGs) catalyze the covalent crosslinking of proteins via isopeptide bonds. The most prominent isoform, TG2, is associated with physiological processes such as extracellular matrix (ECM) stabilization and plays a crucial role in the pathogenesis of e.g. fibrotic diseases, cancer and celiac disease. Therefore, TG2 represents a pharmacological target of increasing relevance. The glycosaminoglycans (GAG) heparin (HE) and heparan sulfate (HS) constitute high-affinity interaction partners of TG2 in the ECM. Chemically modified GAG are promising molecules for pharmacological applications as their composition and chemical functionalization may be used to tackle the function of ECM molecular systems, which has been recently described for hyaluronan (HA) and chondroitin sulfate (CS). Herein, we investigate the recognition of GAG derivatives by TG2 using an enzyme-crosslinking activity assay in combination with in silico molecular modeling and docking techniques. The study reveals that GAG represent potent inhibitors of TG2 crosslinking activity and offers atom-detailed mechanistic insights.
Collapse
Affiliation(s)
- Claudia Damaris Müller
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Gloria Ruiz-Gómez
- Structural Bioinformatics, BIOTEC, Technische Universität Dresden, Tatzberg 47-51, 01307, Dresden, Germany
| | - Sophie Cazzonelli
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Stephanie Möller
- Biomaterials Department, INNOVENT e.V., Prüssingstraße 27 B, 07745, Jena, Germany
| | - Robert Wodtke
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Reik Löser
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Joanna Freyse
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2/4, 14195, Berlin, Germany
| | - Jan-Niklas Dürig
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2/4, 14195, Berlin, Germany
| | - Jörg Rademann
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2/4, 14195, Berlin, Germany
| | - Ute Hempel
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - M Teresa Pisabarro
- Structural Bioinformatics, BIOTEC, Technische Universität Dresden, Tatzberg 47-51, 01307, Dresden, Germany.
| | - Sarah Vogel
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
| |
Collapse
|
21
|
Amundsen SF, Stamnaes J, du Pré MF, Sollid LM. Transglutaminase 2 affinity and enzyme-substrate intermediate stability as determining factors for T-cell responses to gluten peptides in celiac disease. Eur J Immunol 2022; 52:1474-1481. [PMID: 35715890 PMCID: PMC9545004 DOI: 10.1002/eji.202249862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/06/2022] [Accepted: 06/15/2022] [Indexed: 11/18/2022]
Abstract
The adaptive immune response of celiac disease (CeD) involves presentation of gluten peptides to CD4+ T cells by transglutaminase 2 (TG2) specific B cells. This B‐cell/T‐cell crosstalk is facilitated by involvement of TG2:gluten peptide complexes that act principally in the form of enzyme‐substrate intermediates. Here, we have addressed how gluten peptide affinity and complex stability in the presence of secondary substrates affect the uptake of TG2:gluten peptide complexes by TG2‐specific B cells and the activation of gluten‐specific T cells. We studied affinity of various gluten peptides for TG2 by biochemical assay, and monitored uptake of gluten peptides by TG2‐specific B cells by flow cytometry. Crosstalk between TG2‐specific B cells and gluten‐specific T cells was assayed with transfectants expressing antigen receptors derived from CeD patients. We found that gluten peptides with high TG2 affinity showed better uptake by TG2‐specific B cells. Uptake by B cells, and subsequent activation of T cells, was negatively affected by polyamines acting as secondary TG2 substrates. These results show that affinity between gluten peptide and TG2 governs the selection of T‐cell epitopes via enhanced uptake of TG2:gluten complexes by TG2‐specific B cells, and that exogenous polyamines can influence the CeD immune responses by disrupting TG2:gluten complexes.
Collapse
Affiliation(s)
- Sunniva F Amundsen
- KG Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jorunn Stamnaes
- KG Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Marie Fleur du Pré
- KG Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department Immunology, Oslo University Hospital, Oslo, Norway
| | - Ludvig M Sollid
- KG Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
22
|
Kar S, Vu K, Mottamal M, Al-Horani RA. Ethacrynic acid is an inhibitor of human factor XIIIa. BMC Pharmacol Toxicol 2022; 23:35. [PMID: 35642005 PMCID: PMC9158266 DOI: 10.1186/s40360-022-00575-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 05/25/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Ethacrynic acid (EA) is a loop diuretic that is approved orally and parenterally to manage edema-associated diseases. Nevertheless, it was earlier reported that it is also associated with bleeding upon its parenteral administration. In this report, we investigated the effects of EA on human factor XIIIa (FXIIIa) of the coagulation process using a variety of techniques.
Methods
A series of biochemical and computational methods have been used in this study. The potency and efficacy of human FXIIIa inhibition by EA was evaluated using a bisubstrate-based fluorescence trans-glutamination assay under near physiological conditions. To establish the physiological relevance of FXIIIa inhibition by EA, the effect on FXIIIa-mediated polymerization of fibrin(ogen) as well as the formation of fibrin(ogen) – α2-antiplasmin complex was evaluated using SDS-PAGE experiments. The selectivity profile of EA against other coagulation proteins was assessed by evaluating EA’s effect on human clotting times in the activated partial thromboplastin time (APTT) and the prothrombin time (PT) assays. We also used molecular modeling studies to put forward a putative binding mode for EA in the active site of FXIIIa. Results involving EA were the average of at least three experiments and the standard error ± 1 was provided. In determining the inhibition parameters, we used non-linear regression analysis.
Results
FXIIIa is a transglutaminase that works at the end of the coagulation process to form an insoluble, rigid, and cross-linked fibrin rich blood clot. In fact, inhibition of FXIIIa-mediated biological processes has been reported to result in a bleeding diathesis. Inhibition of FXIIIa by EA was investigated given the nucleophilic nature of the thiol-containing active site of the enzyme and the Michael acceptor-based electrophilicity of EA. In a bisubstrate-based fluorescence trans-glutamination assay, EA inhibited FXIIIa with a moderate potency (IC50 ~ 105 µM) and efficacy (∆Y ~ 66%). In SDS-PAGE experiments, EA appears to significantly inhibit the FXIIIa-mediated polymerization of fibrin(ogen) as well as the formation of fibrin(ogen) – α2-antiplasmin complex which indicates that EA affects the physiological functions of FXIIIa. Interestingly, EA did not affect the clotting times of human plasma in the APTT and the PT assays at the highest concentration tested of 2.5 mM suggesting the lack of effects on the coagulation serine proteases and potentially the functional selectivity of EA with respect to the clotting process. Molecular modeling studies demonstrated that the Michael acceptor of EA forms a covalent bond with catalytic residue of Cys314 in the active site of FXIIIa.
Conclusions
Overall, our studies indicate that EA inhibits the physiological function of human FXIIIa in vitro which may potentially contribute to the bleeding complications that were reported with the association of the parenteral administration of EA.
Collapse
|
23
|
Rangaswamy AMM, Navals P, Gates EWJ, Shad S, Watt SKI, Keillor JW. Structure-activity relationships of hydrophobic alkyl acrylamides as tissue transglutaminase inhibitors. RSC Med Chem 2022; 13:413-428. [PMID: 35647547 PMCID: PMC9020614 DOI: 10.1039/d1md00382h] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/25/2022] [Indexed: 01/28/2023] Open
Abstract
Tissue transglutaminase (TG2) is a multifunctional protein that plays biological roles based on its ability to catalyse protein cross-linking and to function as a non-canonical G-protein known as Ghα. The non-regulated activity of TG2 has been implicated in fibrosis, celiac disease and the survival of cancer stem cells, underpinning the therapeutic potential of cell permeable small molecule inhibitors of TG2. In the current study, we designed a small library of inhibitors to explore the importance of a terminal hydrophobic moiety, as well as the length of the tether to the irreversible acrylamide warhead. Subsequent kinetic evaluation using an in vitro activity assay provided values for the k inact and K I parameters for each of these irreversible inhibitors. The resulting structure-activity relationship (SAR) clearly indicated the affinity conferred by dansyl and adamantyl moieties, as well as the efficiency provided by the shortest warhead tether. We also provide the first direct evidence of the capability of these inhibitors to suppress the GTP binding ability of TG2, at least partially. However, it is intriguing to note that the SAR trends observed herein are opposite to those predicted by molecular modelling - namely that longer tether groups should improve binding affinity by allowing for deeper insertion of the hydrophobic moiety into a hydrophobic pocket on the enzyme. This discrepancy leads us to question whether the existing crystallographic structures of TG2 are appropriate for docking non-peptidic inhibitors. In the absence of a more relevant crystallographic structure, the data from rigorous kinetic studies, such as those provided herein, are critically important for the development of future small molecule TG2 inhibitors.
Collapse
Affiliation(s)
- Alana M. M. Rangaswamy
- Department of Chemistry and Biomolecular Sciences, University of OttawaOttawaOntario K1N 6N5Canada
| | - Pauline Navals
- Department of Chemistry and Biomolecular Sciences, University of OttawaOttawaOntario K1N 6N5Canada
| | - Eric W. J. Gates
- Department of Chemistry and Biomolecular Sciences, University of OttawaOttawaOntario K1N 6N5Canada
| | - Sammir Shad
- Department of Chemistry and Biomolecular Sciences, University of OttawaOttawaOntario K1N 6N5Canada
| | - Sarah K. I. Watt
- Department of Chemistry and Biomolecular Sciences, University of OttawaOttawaOntario K1N 6N5Canada
| | - Jeffrey W. Keillor
- Department of Chemistry and Biomolecular Sciences, University of OttawaOttawaOntario K1N 6N5Canada
| |
Collapse
|
24
|
Alavarse AC, Frachini ECG, da Silva RLCG, Lima VH, Shavandi A, Petri DFS. Crosslinkers for polysaccharides and proteins: Synthesis conditions, mechanisms, and crosslinking efficiency, a review. Int J Biol Macromol 2022; 202:558-596. [PMID: 35038469 DOI: 10.1016/j.ijbiomac.2022.01.029] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/20/2021] [Accepted: 01/06/2022] [Indexed: 01/16/2023]
Abstract
Polysaccharides and proteins are important macromolecules for developing hydrogels devoted to biomedical applications. Chemical hydrogels offer chemical, mechanical, and dimensional stability than physical hydrogels due to the chemical bonds among the chains mediated by crosslinkers. There are many crosslinkers to synthesize polysaccharides and proteins based on hydrogels. In this review, we revisited the crosslinking reaction mechanisms between synthetic or natural crosslinkers and polysaccharides or proteins. The selected synthetic crosslinkers were glutaraldehyde, carbodiimide, boric acid, sodium trimetaphosphate, N,N'-methylene bisacrylamide, and polycarboxylic acid, whereas the selected natural crosslinkers included transglutaminase, tyrosinase, horseradish peroxidase, laccase, sortase A, genipin, vanillin, tannic acid, and phytic acid. No less important are the reactions involving click chemistry and the macromolecular crosslinkers for polysaccharides and proteins. Literature examples of polysaccharides or proteins crosslinked by the different strategies were presented along with the corresponding highlights. The general mechanism involved in chemical crosslinking mediated by gamma and UV radiation was discussed, with particular attention to materials commonly used in digital light processing. The evaluation of crosslinking efficiency by gravimetric measurements, rheology, and spectroscopic techniques was presented. Finally, we presented the challenges and opportunities to create safe chemical hydrogels for biomedical applications.
Collapse
Affiliation(s)
- Alex Carvalho Alavarse
- Fundamental Chemistry Department, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Emilli Caroline Garcia Frachini
- Fundamental Chemistry Department, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | | | - Vitoria Hashimoto Lima
- Fundamental Chemistry Department, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Amin Shavandi
- Université libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium
| | - Denise Freitas Siqueira Petri
- Fundamental Chemistry Department, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil.
| |
Collapse
|
25
|
Structure-activity relationships of N-terminal variants of peptidomimetic tissue transglutaminase inhibitors. Eur J Med Chem 2022; 232:114172. [DOI: 10.1016/j.ejmech.2022.114172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/29/2022] [Accepted: 01/30/2022] [Indexed: 02/07/2023]
|
26
|
Rorke EA, Adhikary G, Szmacinski H, Lakowicz JR, Weber DJ, Godoy-Ruiz R, Puranik P, Keillor JW, Gates EW, Eckert RL. Sulforaphane covalently interacts with the transglutaminase 2 cancer maintenance protein to alter its structure and suppress its activity. Mol Carcinog 2022; 61:19-32. [PMID: 34610184 PMCID: PMC8665039 DOI: 10.1002/mc.23356] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 01/03/2023]
Abstract
Type 2 transglutaminase (TG2) functions as an important cancer cell survival protein in a range of cancers including epidermal squamous cell carcinoma. TG2 exists in open and closed conformations each of which has a distinct and mutually exclusive activity. The closed conformation has GTP-binding/GTPase activity while the open conformation functions as a transamidase to catalyze protein-protein crosslinking. GTP-binding/GTPase activity is required for TG2 maintenance of the aggressive cancer phenotype. Thus, identifying agents that convert TG2 from the closed to the open GTP-binding/GTPase inactive conformation is an important cancer prevention/treatment strategy. Sulforaphane (SFN) is an important diet-derived cancer prevention agent that is known to possess a reactive isothiocyanate group and has potent anticancer activity. Using a biotin-tagged SFN analog (Biotin-ITC) and kinetic analysis we show that SFN covalently and irreversibly binds to recombinant TG2 to inhibit transamidase activity and shift TG2 to an open/extended conformation, leading to a partial inhibition of GTP binding. We also show that incubation of cancer cells or cancer cell extract with Biotin-ITC results in formation of a TG2/Biotin-ITC complex and that SFN treatment of cancer cells inhibits TG2 transamidase activity and shifts TG2 to an open/extended conformation. These findings identify TG2 as a direct SFN anticancer target in epidermal squamous cell carcinoma.
Collapse
Affiliation(s)
- Ellen A. Rorke
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - Gautam Adhikary
- Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - Henryk Szmacinski
- Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - Joseph R. Lakowicz
- Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - David J. Weber
- Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, 21201,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, 21201,Center for Biomolecular Therapueutics, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - Raquel Godoy-Ruiz
- Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, 21201,Center for Biomolecular Therapueutics, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - Purushottamachar Puranik
- Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, 21201,Center for Biomolecular Therapueutics, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | | | - Eric W.J Gates
- Department of Chemistry, University of Ottawa, ON, Canada
| | - Richard L. Eckert
- Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, 21201,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, 21201,Center for Biomolecular Therapueutics, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| |
Collapse
|
27
|
Lactoferrin particles assembled via transglutaminase-induced crosslinking: Utilization in oleogel-based Pickering emulsions with improved curcumin bioaccessibility. Food Chem 2021; 374:131779. [PMID: 34896951 DOI: 10.1016/j.foodchem.2021.131779] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 11/22/2022]
Abstract
In this study, the optimal environmental condition for preparation of lactoferrin particles assembled via transglutaminase-induced crosslinking (TG-LF particles) was pH 8, 100 U/g of TG concentration, 50 °C and 2 h of crosslinking time. Contact angle of TG-LF particles was 79°. Then, corn oil-based oleogels were prepared with carnauba wax (CW), behenyl alcohol (BA) and CW-BA mixture at 1:4 ratio (MT). To investigate the effect of oleogels on oleogel-based Pickering emulsions, oleogel-based Pickering emulsions were prepared by a two-step method using different oleogels as the oil phase and the TG-LF particles as the emulsifier. In vitro digestion study revealed that CW oleogel-based Pickering emulsion had the highest lipolysis rate and curcumin bioaccessibility. This study demonstrated that TG-LF particle-stabilized oleogel-based Pickering emulsions had good performance in curcumin delivery, which provided a new idea for the preparation of Pickering emulsifier and enriched the knowledge in the field of oleogel-based Pickering emulsion.
Collapse
|
28
|
Keillor JW, Johnson GVW. Transglutaminase 2 as a therapeutic target for neurological conditions. Expert Opin Ther Targets 2021; 25:721-731. [PMID: 34607527 DOI: 10.1080/14728222.2021.1989410] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/01/2021] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Transglutaminase 2 (TG2) has been implicated in numerous neurological conditions, including neurodegenerative diseases, multiple sclerosis, and CNS injury. Early studies on the role of TG2 in neurodegenerative conditions focused on its ability to 'crosslink' proteins into insoluble aggregates. However, more recent studies have suggested that this is unlikely to be the primary mechanism by which TG2 contributes to the pathogenic processes. Although the specific mechanisms by which TG2 is involved in neurological conditions have not been clearly defined, TG2 regulates numerous cellular processes through which it could contribute to a specific disease. Given the fact that TG2 is a stress-induced gene and elevated in disease or injury conditions, TG2 inhibitors may be useful neurotherapeutics. AREAS COVERED Overview of TG2 and different TG2 inhibitors. A brief review of TG2 in neurodegenerative diseases, multiple sclerosis and CNS injury and inhibitors that have been tested in different models. Database search: https://pubmed.ncbi.nlm.nih.gov prior to 1 July 2021. EXPERT OPINION Currently, it appears unlikely that inhibiting TG2 in the context of neurodegenerative diseases would be therapeutically advantageous. However, for multiple sclerosis and CNS injuries, TG2 inhibitors may have the potential to be therapeutically useful and thus there is rationale for their further development.
Collapse
Affiliation(s)
- Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Gail V W Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, USA
| |
Collapse
|
29
|
Pehrsson M, Mortensen JH, Manon-Jensen T, Bay-Jensen AC, Karsdal MA, Davies MJ. Enzymatic cross-linking of collagens in organ fibrosis - resolution and assessment. Expert Rev Mol Diagn 2021; 21:1049-1064. [PMID: 34330194 DOI: 10.1080/14737159.2021.1962711] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Enzymatic cross-linking of the collagens within the extracellular matrix (ECM) catalyzed by enzymes such as lysyl oxidase (LOX) and lysyl oxidase like-enzymes 1-4 (LOXL), transglutaminase 2 (TG2), and peroxidasin (PXDN) contribute to fibrosis progression through extensive collagen cross-linking. Studies in recent years have begun elucidating the important role of collagen cross-linking in perpetuating progression of organ fibrosis independently of inflammation through an increasingly stiff and noncompliant ECM. Therefore, collagen cross-linking and the cross-linking enzymes have become new targets in anti-fibrotic therapy as well as targets of novel biomarkers to properly assess resolution of the fibrotic ECM.Areas covered: The enzymatic actions of enzymes catalyzing collagen cross-linking and their relevance in organ fibrosis. Potential biomarkers specifically quantifying proteolytic fragments of collagen cross-linking is discussed based on Pubmed search done in November 2020 as well as the authors knowledge.Expert opinion: Current methods for the assessment of fibrosis involve the use of invasive and/or cumbersome and expensive methods such as tissue biopsies. Thus, an unmet need exists for the development and validation of minimally invasive biomarkers of proteolytic fragments of cross-linked collagens. These biomarkers may aid in the development and proper assessment of fibrosis resolution in coming years.
Collapse
Affiliation(s)
- Martin Pehrsson
- Department of Biomedical Science, University of Copenhagen, Copenhagen, Denmark.,Biomarkers & Research, Nordic Bioscience A/S, Herlev, Denmark
| | | | | | | | | | | |
Collapse
|
30
|
Tatsukawa H, Hitomi K. Role of Transglutaminase 2 in Cell Death, Survival, and Fibrosis. Cells 2021; 10:cells10071842. [PMID: 34360011 PMCID: PMC8307792 DOI: 10.3390/cells10071842] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/27/2022] Open
Abstract
Transglutaminase 2 (TG2) is a ubiquitously expressed enzyme catalyzing the crosslinking between Gln and Lys residues and involved in various pathophysiological events. Besides this crosslinking activity, TG2 functions as a deamidase, GTPase, isopeptidase, adapter/scaffold, protein disulfide isomerase, and kinase. It also plays a role in the regulation of hypusination and serotonylation. Through these activities, TG2 is involved in cell growth, differentiation, cell death, inflammation, tissue repair, and fibrosis. Depending on the cell type and stimulus, TG2 changes its subcellular localization and biological activity, leading to cell death or survival. In normal unstressed cells, intracellular TG2 exhibits a GTP-bound closed conformation, exerting prosurvival functions. However, upon cell stimulation with Ca2+ or other factors, TG2 adopts a Ca2+-bound open conformation, demonstrating a transamidase activity involved in cell death or survival. These functional discrepancies of TG2 open form might be caused by its multifunctional nature, the existence of splicing variants, the cell type and stimulus, and the genetic backgrounds and variations of the mouse models used. TG2 is also involved in the phagocytosis of dead cells by macrophages and in fibrosis during tissue repair. Here, we summarize and discuss the multifunctional and controversial roles of TG2, focusing on cell death/survival and fibrosis.
Collapse
|
31
|
Song W, Ko J, Choi YH, Hwang NS. Recent advancements in enzyme-mediated crosslinkable hydrogels: In vivo-mimicking strategies. APL Bioeng 2021; 5:021502. [PMID: 33834154 PMCID: PMC8018798 DOI: 10.1063/5.0037793] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/03/2021] [Indexed: 12/19/2022] Open
Abstract
Enzymes play a central role in fundamental biological processes and have been traditionally used to trigger various processes. In recent years, enzymes have been used to tune biomaterial responses and modify the chemical structures at desired sites. These chemical modifications have allowed the fabrication of various hydrogels for tissue engineering and therapeutic applications. This review provides a comprehensive overview of recent advancements in the use of enzymes for hydrogel fabrication. Strategies to enhance the enzyme function and improve biocompatibility are described. In addition, we describe future opportunities and challenges for the production of enzyme-mediated crosslinkable hydrogels.
Collapse
Affiliation(s)
- Wonmoon Song
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Junghyeon Ko
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Hwan Choi
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Nathaniel S. Hwang
- Author to whom correspondence should be addressed:. Tel.: 82-2-880-1635. Fax: 82-2-880-7295
| |
Collapse
|
32
|
Aberoumand A, Baesi F. The Effects of Surimi Process on Amino Acids Profile of Sphyraena jello Fish. JOURNAL OF AQUATIC FOOD PRODUCT TECHNOLOGY 2021. [DOI: 10.1080/10498850.2021.1882634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Ali Aberoumand
- Department of Fisheries, Behbahan Khatam Alanbia University of Technology, Behbahan, Iran
| | - Ferideh Baesi
- MSc in Fisheries Department, Behbahan Khatam Alanbia University of Technology, Behbahan, Iran
| |
Collapse
|
33
|
Wodtke R, Wodtke J, Hauser S, Laube M, Bauer D, Rothe R, Neuber C, Pietsch M, Kopka K, Pietzsch J, Löser R. Development of an 18F-Labeled Irreversible Inhibitor of Transglutaminase 2 as Radiometric Tool for Quantitative Expression Profiling in Cells and Tissues. J Med Chem 2021; 64:3462-3478. [PMID: 33705656 DOI: 10.1021/acs.jmedchem.1c00096] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The transamidase activity of transglutaminase 2 (TGase 2) is considered to be important for several pathophysiological processes including fibrotic and neoplastic tissue growth, whereas in healthy cells this enzymatic function is predominantly latent. Methods that enable the highly sensitive detection of TGase 2, such as application of radiolabeled activity-based probes, will support the exploration of the enzyme's function in various diseases. In this context, the radiosynthesis and detailed in vitro radiopharmacological evaluation of an 18F-labeled Nε-acryloyllysine piperazide are reported. Robust and facile detection of the radiotracer-TGase 2 complex by autoradiography of thin layer plates and polyacrylamide gels after chromatographic and electrophoretic separation owing to irreversible covalent bond formation was demonstrated for the isolated protein, cell lysates, and living cells. By use of this radiotracer, quantitative data on the expression profile of activatable TGase 2 in mouse organs and selected tumors were obtained for the first time by autoradiography of tissue sections.
Collapse
Affiliation(s)
- Robert Wodtke
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Johanna Wodtke
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Sandra Hauser
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Markus Laube
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - David Bauer
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Fakultät Chemie und Lebensmittelchemie, Technische Universität Dresden, Mommsenstraße 4, 01062 Dresden, Germany
| | - Rebecca Rothe
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Fakultät Chemie und Lebensmittelchemie, Technische Universität Dresden, Mommsenstraße 4, 01062 Dresden, Germany
| | - Christin Neuber
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Markus Pietsch
- Institut II für Pharmakologie, Zentrum für Pharmakologie, Medizinische Fakultät, Universität zu Köln, Gleueler Straße 24, 50931 Köln, Germany
| | - Klaus Kopka
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Fakultät Chemie und Lebensmittelchemie, Technische Universität Dresden, Mommsenstraße 4, 01062 Dresden, Germany
| | - Jens Pietzsch
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Fakultät Chemie und Lebensmittelchemie, Technische Universität Dresden, Mommsenstraße 4, 01062 Dresden, Germany
| | - Reik Löser
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Fakultät Chemie und Lebensmittelchemie, Technische Universität Dresden, Mommsenstraße 4, 01062 Dresden, Germany
| |
Collapse
|
34
|
Crosslinking Collagen Constructs: Achieving Cellular Selectivity Through Modifications of Physical and Chemical Properties. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10196911] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Collagen-based constructs have emerged in recent years as ideal candidates for tissue engineering implants. For many biomedical applications, collagen is crosslinked in order to improve the strength, stiffness and stability of the construct. However, the crosslinking process may also result in unintended changes to cell viability, adhesion or proliferation on the treated structures. This review provides a brief overview of some of both the most commonly used and novel crosslinkers used with collagen, and suggests a framework by which crosslinking methods can be compared and selected for a given tissue engineering application.
Collapse
|
35
|
Al-Horani RA, Kar S. Factor XIIIa inhibitors as potential novel drugs for venous thromboembolism. Eur J Med Chem 2020; 200:112442. [PMID: 32502864 PMCID: PMC7513741 DOI: 10.1016/j.ejmech.2020.112442] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/11/2022]
Abstract
Human factor XIIIa (FXIIIa) is a multifunctional transglutaminase with a significant role in hemostasis. FXIIIa catalyzes the last step in the coagulation process. It stabilizes the blood clot by cross-linking the α- and γ-chains of fibrin. It also protects the newly formed clot from plasmin-mediated fibrinolysis, primarily by cross-linking α2-antiplasmin to fibrin. Furthermore, FXIIIa is a major determinant of clot size and clot's red blood cells content. Therefore, inhibitors targeting FXIIIa have been considered to develop a new generation of anticoagulants to prevent and/or treat venous thromboembolism. Several inhibitors of FXIIIa have been discovered or designed including active site and allosteric site small molecule inhibitors as well as natural and modified polypeptides. This work reviews the structural, biochemical, and pharmacological aspects of FXIIIa inhibitors so as to advance their molecular design to become more clinically relevant.
Collapse
Affiliation(s)
- Rami A Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA, 70125, USA.
| | - Srabani Kar
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA, 70125, USA
| |
Collapse
|
36
|
Moonlighting Proteins at the Candidal Cell Surface. Microorganisms 2020; 8:microorganisms8071046. [PMID: 32674422 PMCID: PMC7409194 DOI: 10.3390/microorganisms8071046] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/11/2020] [Accepted: 07/12/2020] [Indexed: 12/31/2022] Open
Abstract
The cell wall in Candida albicans is not only a tight protective envelope but also a point of contact with the human host that provides a dynamic response to the constantly changing environment in infection niches. Particularly important roles are attributed to proteins exposed at the fungal cell surface. These include proteins that are stably and covalently bound to the cell wall or cell membrane and those that are more loosely attached. Interestingly in this regard, numerous loosely attached proteins belong to the class of “moonlighting proteins” that are originally intracellular and that perform essentially different functions in addition to their primary housekeeping roles. These proteins also demonstrate unpredicted interactions with non-canonical partners at an a priori unexpected extracellular location, achieved via non-classical secretion routes. Acting both individually and collectively, the moonlighting proteins contribute to candidal virulence and pathogenicity through their involvement in mechanisms critical for successful host colonization and infection, such as the adhesion to host cells, interactions with plasma homeostatic proteolytic cascades, responses to stress conditions and molecular mimicry. The documented knowledge of the roles of these proteins in C. albicans pathogenicity has utility for assisting the design of new therapeutic, diagnostic and preventive strategies against candidiasis.
Collapse
|
37
|
Maddock RMA, Pollard GJ, Moreau NG, Perry JJ, Race PR. Enzyme-catalysed polymer cross-linking: Biocatalytic tools for chemical biology, materials science and beyond. Biopolymers 2020; 111:e23390. [PMID: 32640085 DOI: 10.1002/bip.23390] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022]
Abstract
Intermolecular cross-linking is one of the most important techniques that can be used to fundamentally alter the material properties of a polymer. The introduction of covalent bonds between individual polymer chains creates 3D macromolecular assemblies with enhanced mechanical properties and greater chemical or thermal tolerances. In contrast to many chemical cross-linking reactions, which are the basis of thermoset plastics, enzyme catalysed processes offer a complimentary paradigm for the assembly of cross-linked polymer networks through their predictability and high levels of control. Additionally, enzyme catalysed reactions offer an inherently 'greener' and more biocompatible approach to covalent bond formation, which could include the use of aqueous solvents, ambient temperatures, and heavy metal-free reagents. Here, we review recent progress in the development of biocatalytic methods for polymer cross-linking, with a specific focus on the most promising candidate enzyme classes and their underlying catalytic mechanisms. We also provide exemplars of the use of enzyme catalysed cross-linking reactions in industrially relevant applications, noting the limitations of these approaches and outlining strategies to mitigate reported deficiencies.
Collapse
Affiliation(s)
- Rosie M A Maddock
- School of Biochemistry, University of Bristol, University Walk, Bristol, UK.,BrisSynBio Synthetic Biology Research Centre, Life Sciences Building, Tyndall Avenue University of Bristol, Bristol, UK
| | - Gregory J Pollard
- School of Biochemistry, University of Bristol, University Walk, Bristol, UK
| | - Nicolette G Moreau
- School of Biochemistry, University of Bristol, University Walk, Bristol, UK
| | - Justin J Perry
- Department of Applied Sciences, Northumbria University, Ellison Building, Newcastle upon Tyne, UK
| | - Paul R Race
- School of Biochemistry, University of Bristol, University Walk, Bristol, UK.,BrisSynBio Synthetic Biology Research Centre, Life Sciences Building, Tyndall Avenue University of Bristol, Bristol, UK
| |
Collapse
|
38
|
Kang JH, Lee SH, Lee JS, Oh SJ, Ha JS, Choi HJ, Kim SY. Inhibition of Transglutaminase 2 but Not of MDM2 Has a Significant Therapeutic Effect on Renal Cell Carcinoma. Cells 2020; 9:cells9061475. [PMID: 32560270 PMCID: PMC7349864 DOI: 10.3390/cells9061475] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023] Open
Abstract
More than 50% of human cancers harbor TP53 mutations and increased expression of Mouse double minute 2 homolog(MDM2), which contribute to cancer progression and drug resistance. Renal cell carcinoma (RCC) has an unusually high incidence of wild-type p53, with a mutation rate of less than 4%. MDM2 is master regulator of apoptosis in cancer cells, which is triggered through proteasomal degradation of wild-type p53. Recently, we found that p53 protein levels in RCC are regulated by autophagic degradation. Transglutaminase 2 (TGase 2) was responsible for p53 degradation through this pathway. Knocking down TGase 2 increased p53-mediated apoptosis in RCC. Therefore, we asked whether depleting p53 from RCC cells occurs via MDM2-mediated proteasomal degradation or via TGase 2-mediated autophagic degradation. In vitro gene knockdown experiments revealed that stability of p53 in RCC was inversely related to levels of both MDM2 and TGase 2 protein. Therefore, we examined the therapeutic efficacy of inhibitors of TGase 2 and MDM2 in an in vivo model of RCC. The results showed that inhibiting TGase 2 but not MDM2 had efficient anticancer effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Soo-Youl Kim
- Correspondence: ; Tel.: +82-31-920-2221; Fax: +82-31-920-2278
| |
Collapse
|
39
|
Tunable bioactivity and mechanics of collagen-based tissue engineering constructs: A comparison of EDC-NHS, genipin and TG2 crosslinkers. Biomaterials 2020; 254:120109. [PMID: 32480093 PMCID: PMC7298615 DOI: 10.1016/j.biomaterials.2020.120109] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/24/2020] [Accepted: 05/07/2020] [Indexed: 02/05/2023]
Abstract
Due to its ubiquity and versatility in the human body, collagen is an ideal base material for tissue-engineering constructs. Chemical crosslinking treatments allow precise control of the biochemical and mechanical properties through macromolecular modifications to the structure of collagen. In this work, three key facets regarding the collagen crosslinking process are explored. Firstly, a comparison is drawn between the carbodiimide-succinimide (EDC-NHS) system and two emerging crosslinkers utilising alternate chemistries: genipin and tissue transglutaminase (TG2). By characterising the chemical changes upon treatment, the effect of EDC-NHS, genipin and TG2 crosslinking mechanisms on the chemical structure of collagen, and thus the mechanical properties conferred to the substrate is explored. Secondly, the relative importance of mechanical and biochemical cues on cellular phenomena are investigated, including cell viability, integrin-specific attachment, spreading and proliferation. Here, we observe that for human dermal fibroblasts, long-term, stable proliferation is preconditioned by the availability of suitable binding sites, irrespective of the substrate modulus post-crosslinking. Finally, as seen in the graphical abstract we show that by choosing the appropriate crosslinker chemistries, a materials selection map can be drawn for collagen films, encompassing both a range of tensile modulus and fibroblast proliferation which can be modified independently. Thus, in addition to a range of parameters that can be modified in collagen constructs, we demonstrate a route to obtaining tunable bioactivity and mechanics in collagen constructs is uncovered, that is exclusively driven by the crosslinking process.
Collapse
|
40
|
Mu Y, Wang Y, Huang Y, Li D, Han Y, Chang M, Fu J, Xie Y, Ren J, Wang H, Zhang Y, Luo ZQ, Feng Y. Structural insights into the mechanism and inhibition of transglutaminase-induced ubiquitination by the Legionella effector MavC. Nat Commun 2020; 11:1774. [PMID: 32286321 PMCID: PMC7156659 DOI: 10.1038/s41467-020-15645-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/20/2020] [Indexed: 01/09/2023] Open
Abstract
Protein ubiquitination is one of the most prevalent post-translational modifications, controlling virtually every process in eukaryotic cells. Recently, the Legionella effector MavC was found to mediate a unique ubiquitination through transglutamination, linking ubiquitin (Ub) to UBE2N through UbGln40 in a process that can be inhibited by another Legionella effector, Lpg2149. Here, we report the structures of MavC/UBE2N/Ub ternary complex, MavC/UBE2N-Ub (product) binary complex, and MavC/Lpg2149 binary complex. During the ubiquitination, the loop containing the modification site K92 of UBE2N undergoes marked conformational change, and Lpg2149 inhibits this ubiquitination through competing with Ub to bind MavC. Moreover, we found that MavC itself also exhibits weak deubiquitinase activity towards this non-canonical ubiquitination. Together, our study not only provides insights into the mechanism and inhibition of this transglutaminase-induced ubiquitination by MavC, but also sheds light on the future studies into UBE2N inhibition by this modification and deubiquitinases of this unique ubiquitination.
Collapse
Affiliation(s)
- Yajuan Mu
- 0000 0000 9931 8406grid.48166.3dBeijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, 100029 Beijing, China
| | - Yue Wang
- 0000 0000 9931 8406grid.48166.3dBeijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, 100029 Beijing, China
| | - Yanfei Huang
- 0000 0000 9931 8406grid.48166.3dBeijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, 100029 Beijing, China
| | - Dong Li
- 0000 0000 9931 8406grid.48166.3dBeijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, 100029 Beijing, China
| | - Youyou Han
- 0000 0000 9931 8406grid.48166.3dBeijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, 100029 Beijing, China
| | - Min Chang
- 0000 0000 9931 8406grid.48166.3dBeijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, 100029 Beijing, China
| | - Jiaqi Fu
- 0000 0004 1937 2197grid.169077.ePurdue Institute for Inflammation, Immunology and Infectious Disease and Department of Biological Sciences, Purdue University, West Lafayette, IN USA
| | - Yongchao Xie
- 0000 0000 9931 8406grid.48166.3dBeijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, 100029 Beijing, China
| | - Jie Ren
- 0000 0001 0526 1937grid.410727.7State Key Laboratory for Biology of Plant Diseases and Insect Pests/Key Laboratory of Control of Biological Hazard Factors (Plant Origin) for Agri-product Quality and Safety, Ministry of Agriculture, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, 100081 Beijing, China
| | - Hao Wang
- 0000 0000 9931 8406grid.48166.3dBeijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, 100029 Beijing, China
| | - Yi Zhang
- 0000 0000 9931 8406grid.48166.3dBeijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, 100029 Beijing, China
| | - Zhao-Qing Luo
- 0000 0004 1937 2197grid.169077.ePurdue Institute for Inflammation, Immunology and Infectious Disease and Department of Biological Sciences, Purdue University, West Lafayette, IN USA
| | - Yue Feng
- 0000 0000 9931 8406grid.48166.3dBeijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, 100029 Beijing, China
| |
Collapse
|
41
|
A Precision Strategy to Cure Renal Cell Carcinoma by Targeting Transglutaminase 2. Int J Mol Sci 2020; 21:ijms21072493. [PMID: 32260198 PMCID: PMC7177245 DOI: 10.3390/ijms21072493] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/29/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
In a recent report, no significance of transglutaminase 2 (TGase 2) was noted in the analyses of expression differences between normal and clear cell renal cell carcinoma (ccRCC), although we found that knock down of TGase 2 induced significant p53-mediated cell death in ccRCC. Generally, to find effective therapeutic targets, we need to identify targets that belong specifically to a cancer phenotype that can be differentiated from a normal phenotype. Here, we offer precise reasons why TGase 2 may be the first therapeutic target for ccRCC, according to several lines of evidence. TGase 2 is negatively regulated by von Hippel-Lindau tumor suppressor protein (pVHL) and positively regulated by hypoxia-inducible factor 1-α (HIF-1α) in renal cell carcinoma (RCC). Therefore, most of ccRCC presents high level expression of TGase 2 because over 90% of ccRCC showed VHL inactivity through mutation and methylation. Cell death, angiogenesis and drug resistance were specifically regulated by TGase 2 through p53 depletion in ccRCC because over 90% of ccRCC express wild type p53, which is a cell death inducer as well as a HIF-1α suppressor. Although there have been no detailed studies of the physiological role of TGase 2 in multi-omics analyses of ccRCC, a life-long study of the physiological roles of TGase 2 led to the discovery of the first target as well as the first therapeutic treatment for ccRCC in the clinical field.
Collapse
|
42
|
Solution-phase synthesis of the fluorogenic TGase 2 acyl donor Z-Glu(HMC)-Gly-OH and its use for inhibitor and amine substrate characterisation. Anal Biochem 2020; 595:113612. [DOI: 10.1016/j.ab.2020.113612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 11/20/2022]
|
43
|
Abstract
Protein semisynthesis-defined herein as the assembly of a protein from a combination of synthetic and recombinant fragments-is a burgeoning field of chemical biology that has impacted many areas in the life sciences. In this review, we provide a comprehensive survey of this area. We begin by discussing the various chemical and enzymatic methods now available for the manufacture of custom proteins containing noncoded elements. This section begins with a discussion of methods that are more chemical in origin and ends with those that employ biocatalysts. We also illustrate the commonalities that exist between these seemingly disparate methods and show how this is allowing for the development of integrated chemoenzymatic methods. This methodology discussion provides the technical foundation for the second part of the review where we cover the great many biological problems that have now been addressed using these tools. Finally, we end the piece with a short discussion on the frontiers of the field and the opportunities available for the future.
Collapse
Affiliation(s)
| | - Tom W. Muir
- Department of Chemistry, Princeton University, Frick Laboratory, Princeton, New Jersey 08544, United States
| |
Collapse
|
44
|
Zhang Y, Simpson BK. Food-related transglutaminase obtained from fish/shellfish. Crit Rev Food Sci Nutr 2019; 60:3214-3232. [DOI: 10.1080/10408398.2019.1681357] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Yi Zhang
- Department of Food Science and Agricultural Chemistry, McGill University, Québec, Québec, Canada
| | - Benjamin K. Simpson
- Department of Food Science and Agricultural Chemistry, McGill University, Québec, Québec, Canada
| |
Collapse
|
45
|
Anokhin BA, Dean WL, Smith KA, Flick MJ, Ariëns RAS, Philippou H, Maurer MC. Proteolytic and nonproteolytic activation mechanisms result in conformationally and functionally different forms of coagulation factor XIII A. FEBS J 2019; 287:452-464. [PMID: 31407850 DOI: 10.1111/febs.15040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/21/2019] [Accepted: 08/12/2019] [Indexed: 12/17/2022]
Abstract
Factor XIIIA (FXIIIA) is a transglutaminase that cross-links intra- and extracellular protein substrates. FXIIIA is expressed as an inactive zymogen, and during blood coagulation, it is activated by removal of an activation peptide by the protease thrombin. No such proteolytic FXIIIA activation is known to occur in other tissues or the intracellular form of FXIIIA. For those locations, FXIIIA is assumed instead to undergo activation by Ca2+ ions. Previously, we demonstrated a monomeric state for active FXIIIA. Current analytical ultracentrifugation and kinetic experiments revealed that thrombin-activated FXIIIA has a higher conformational flexibility and a stronger affinity toward glutamine substrate than does nonproteolytically activated FXIIIA. The proteolytic activation of FXIIIA was further investigated in a context of fibrin clotting. In a series of fibrin cross-linking assays and scanning electron microscopy studies of plasma clots, the activation rates of FXIIIA V34X variants were correlated with the extent of fibrin cross-linking and incorporation of nonfibrous protein into the clot. Overall, the results suggest conformational and functional differences between active FXIIIA forms, thus expanding the understanding of FXIIIA function. Those differences may serve as a basis for developing therapeutic strategies to target FXIIIA in different physiological environments. ENZYMES: Factor XIIIA ( EC 2.3.2.13).
Collapse
Affiliation(s)
| | - William L Dean
- Brown Cancer Center, University of Louisville School of Medicine, KY, USA.,Department of Medicine, University of Louisville, KY, USA.,Department of Biochemistry and Molecular Genetics, University of Louisville, KY, USA
| | - Kerrie A Smith
- Leeds Thrombosis Collective, Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, UK
| | - Matthew J Flick
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Robert A S Ariëns
- Leeds Thrombosis Collective, Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, UK
| | - Helen Philippou
- Leeds Thrombosis Collective, Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, UK
| | | |
Collapse
|
46
|
Nair M, Calahorra Y, Kar-Narayan S, Best SM, Cameron RE. Self-assembly of collagen bundles and enhanced piezoelectricity induced by chemical crosslinking. NANOSCALE 2019; 11:15120-15130. [PMID: 31369017 PMCID: PMC7745105 DOI: 10.1039/c9nr04750f] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/15/2019] [Indexed: 05/26/2023]
Abstract
The piezoelectricity of collagen is purported to be linked to many biological processes including bone formation and wound healing. Although the piezoelectricity of tissue-derived collagen has been documented across the length scales, little work has been undertaken to characterise the local electromechanical properties of processed collagen, which is used as a base for tissue-engineering implants. In this work, three chemically distinct treatments used to form structurally and mechanically stable scaffolds-EDC-NHS, genipin and tissue transglutaminase-are investigated for their effect on collagen piezolectricity. Crosslinking with EDC-NHS is noted to produce a distinct self-assembly of the fibres into bundles roughly 300 nm in width regardless of the collagen origin. These fibre bundles also show a localised piezoelectric response, with enhanced vertical piezoelectricity of collagen. Such topographical features are not observed with the other two chemical treatments, although the shear piezoelectric response is significantly enhanced upon crosslinking. These observations are reconciled by a proposed effect of the crosslinking mechanisms on the molecular and nanostructure of collagen. These results highlight the ability to modify the electromechanical properties of collagen using chemical crosslinking methods.
Collapse
Affiliation(s)
- Malavika Nair
- University of Cambridge, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge, CB3 0DS, UK,
| | - Yonatan Calahorra
- University of Cambridge, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge, CB3 0DS, UK,
| | - Sohini Kar-Narayan
- University of Cambridge, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge, CB3 0DS, UK,
| | - Serena M. Best
- University of Cambridge, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge, CB3 0DS, UK,
| | - Ruth E. Cameron
- University of Cambridge, Department of Materials Science and Metallurgy, 27 Charles Babbage Road, Cambridge, CB3 0DS, UK,
| |
Collapse
|
47
|
Balogh G, Muszbek L, Komáromi I. First Step of the Transglutaminase Reaction Catalyzed by Activated Factor XIII Subunit A, Hybrid Quantum Chemistry/Molecular Mechanics Calculations. J Phys Chem B 2019; 123:3887-3897. [DOI: 10.1021/acs.jpcb.9b00542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Gábor Balogh
- Division of Clinical Laboratory Sciences, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary
| | - László Muszbek
- Division of Clinical Laboratory Sciences, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary
| | - István Komáromi
- Division of Clinical Laboratory Sciences, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary
| |
Collapse
|
48
|
Hopkins EJ, Hucl P, Scanlon MG, Nickerson MT. Effect of enzymatic crosslinking on the handling properties of dough as a function of NaCl levels for CWRS varieties, Pembina and Harvest. J Texture Stud 2019; 50:350-358. [PMID: 30861117 DOI: 10.1111/jtxs.12399] [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: 09/04/2018] [Revised: 02/13/2019] [Accepted: 02/28/2019] [Indexed: 11/30/2022]
Abstract
The effects of transglutaminase (TG) and glucose oxidase (GO) on the handling properties of model bread dough were examined at both normal (2% wt. by flour) and reduced (1% wt.) sodium chloride (NaCl) levels using two Canada Western Red Spring (CWRS) cultivars; Pembina and Harvest. The reduction of NaCl level had negative effects on dough rheology and stickiness, however, the inclusion of GO (0.001 and 0.01% by flour weight) or TG (only at the 0.5% by flour weight inclusion) was able to improve dough strength and reduce stickiness. GO appeared to be more effective than TG (at 0.01%) at equivalent concentrations for improving dough-handling properties. Flour cultivar had significant effects; Harvest flour (weaker) was more impacted by salt reduction and enzyme inclusion compared to Pembina flour (stronger). Crosslinking assays showed significant differences in glutenin macropolymer (GMP) content in dough prepared with GO, and dough prepared with different flours. Additionally, significantly fewer free thiol groups were found in dough produced with GO compared to dough without any enzymes and those with TG. GO appears to have potential for use as a bread improver to reduce stickiness and improve the strength of bread dough produced at lower salt concentrations, especially for dough prepared with weaker flour cultivars.
Collapse
Affiliation(s)
- Erin J Hopkins
- Department of Food and Bioproduct Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Pierre Hucl
- Crop Development Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Martin G Scanlon
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Michael T Nickerson
- Department of Food and Bioproduct Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
49
|
The Role of Tissue Transglutaminase in Cancer Cell Initiation, Survival and Progression. Med Sci (Basel) 2019; 7:medsci7020019. [PMID: 30691081 PMCID: PMC6409630 DOI: 10.3390/medsci7020019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/21/2019] [Accepted: 01/22/2019] [Indexed: 12/22/2022] Open
Abstract
Tissue transglutaminase (transglutaminase type 2; TG2) is the most ubiquitously expressed member of the transglutaminase family (EC 2.3.2.13) that catalyzes specific post-translational modifications of proteins through a calcium-dependent acyl-transfer reaction (transamidation). In addition, this enzyme displays multiple additional enzymatic activities, such as guanine nucleotide binding and hydrolysis, protein kinase, disulfide isomerase activities, and is involved in cell adhesion. Transglutaminase 2 has been reported as one of key enzymes that is involved in all stages of carcinogenesis; the molecular mechanisms of action and physiopathological effects depend on its expression or activities, cellular localization, and specific cancer model. Since it has been reported as both a potential tumor suppressor and a tumor-promoting factor, the role of this enzyme in cancer is still controversial. Indeed, TG2 overexpression has been frequently associated with cancer stem cells’ survival, inflammation, metastatic spread, and drug resistance. On the other hand, the use of inducers of TG2 transamidating activity seems to inhibit tumor cell plasticity and invasion. This review covers the extensive and rapidly growing field of the role of TG2 in cancer stem cells survival and epithelial–mesenchymal transition, apoptosis and differentiation, and formation of aggressive metastatic phenotypes.
Collapse
|
50
|
Legionella pneumophila inhibits immune signalling via MavC-mediated transglutaminase-induced ubiquitination of UBE2N. Nat Microbiol 2018; 4:134-143. [PMID: 30420781 PMCID: PMC6294664 DOI: 10.1038/s41564-018-0282-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 10/03/2018] [Indexed: 01/16/2023]
Abstract
The bacterial pathogen Legionella pneumophila modulates host immunity using effectors translocated by its Dot/Icm transporter to facilitate its intracellular replication. A number of these effectors employ diverse mechanisms to interfere with protein ubiquitination, a posttranslational modification essential for immunity. Here we found that L. pneumophila induces monoubiquitination of the E2 enzyme UBE2N by its Dot/Icm substrate MavC(Lpg2147). We demonstrate that MavC is a transglutaminase that catalyzes covalent linkage of ubiquitin to Lys92 and Lys94 of UBE2N via Gln40. Similar to canonical transglutaminases, MavC possess deamidase activity that targets ubiquitin at Gln40. We identified Cys74 as the catalytic residue for both ubiquitination and deamidation activities. Furthermore, ubiquitination of UBE2N by MavC abolishes its activity in the formation of K63-type polyubiquitin chains, which dampens NFκB signaling in the initial phase of bacterial infection. Our results reveal an unprecedented mechanism of modulating host immunity by modifying a key ubiquitination enzyme by ubiquitin transglutamination.
Collapse
|