1
|
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
|
2
|
Cole AG, Kultgen SG, Mani N, Quintero JG, Yi Fan K, Ardzinski A, Stever K, Dorsey BD, Phelps JR, Lee ACH, Thi EP, Chiu T, Tang S, Horanyi PS, Mayclin SJ, Harasym TO, Sofia MJ. Design, synthesis, and structure-activity relationship of a bicyclic HBV capsid assembly modulator chemotype leading to the identification of clinical candidate AB-506. Bioorg Med Chem Lett 2023; 94:129456. [PMID: 37633618 DOI: 10.1016/j.bmcl.2023.129456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/15/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Disruption of the HBV capsid assembly process through small-molecule interaction with HBV core protein is a validated target for the suppression of hepatitis B viral replication and the development of new antivirals. Through combination of key structural features associated with two distinct series of capsid assembly modulators, a novel aminochroman-based chemotype was identified. Optimization of anti-HBV potency through generation of SAR in addition to further core modifications provided a series of related functionalized aminoindanes. Key compounds demonstrated excellent cellular potency in addition to favorable ADME and pharmacokinetic profiles and were shown to be highly efficacious in a mouse model of HBV replication. Aminoindane derivative AB-506 was subsequently advanced into clinical development.
Collapse
Affiliation(s)
- Andrew G Cole
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA.
| | - Steven G Kultgen
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| | - Nagraj Mani
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| | - Jorge G Quintero
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| | - Kristi Yi Fan
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| | - Andrzej Ardzinski
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| | - Kim Stever
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| | - Bruce D Dorsey
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| | - Janet R Phelps
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| | - Amy C H Lee
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| | - Emily P Thi
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| | - Tim Chiu
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| | - Sunny Tang
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| | - Peter S Horanyi
- UCB Pharma, 87 Cambridge Park Drive, Cambridge, MA 02140, USA
| | | | - Troy O Harasym
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| | - Michael J Sofia
- Arbutus Biopharma, Inc., 701 Veterans Circle, Warminster, PA 18974, USA
| |
Collapse
|
3
|
Loppinet E, Besser HA, Lee CE, Zhang W, Cui B, Khosla C. Targeted Lysosomal Degradation of Secreted and Cell Surface Proteins through the LRP-1 Pathway. J Am Chem Soc 2023; 145:18705-18710. [PMID: 37590164 PMCID: PMC10809789 DOI: 10.1021/jacs.3c05109] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Protein dysregulation has been characterized as the cause of pathogenesis in many different diseases. For proteins lacking easily druggable pockets or catalytically active sites, targeted protein degradation is an attractive therapeutic approach. While several methods for targeted protein degradation have been developed, there remains a demand for lower molecular weight molecules that promote efficient degradation of their targets. In this work, we describe the synthesis and validation of a series of heterobifunctional molecules that bind a protein of interest through a small molecule ligand while targeting them to the lysosome using a short gluten peptide that leverages the TG2/LRP-1 pathway. We demonstrate that this approach can be used to effectively endocytose and degrade representative secreted, cell surface, and transmembrane proteins, notably streptavidin, the vitamin B12 receptor, cubilin, and integrin αvβ5. Optimization of these prototypical molecules could generate pharmacologically relevant LYTAC agents.
Collapse
Affiliation(s)
- Elise Loppinet
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Harrison A Besser
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Christina E Lee
- Biophysics Program, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Wei Zhang
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Chaitan Khosla
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan ChEM-H, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
4
|
Valdivia A, Vagadia PP, Guo G, O'Brien E, Matei D, Schiltz GE. Discovery and Characterization of PROTACs Targeting Tissue Transglutaminase (TG2). J Med Chem 2023. [PMID: 37449845 PMCID: PMC10388319 DOI: 10.1021/acs.jmedchem.2c01859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Tissue transglutaminase (TG2) is a multifunctional enzyme involved in the cross-linking of extracellular matrix proteins, formation of complexes with fibronectin (FN) and integrins, and GTP hydrolysis. TG2 is activated in several pathological conditions, including cancer. We recently described a novel series of ligands that bind to TG2 and inhibit its interaction with FN. Because TG2 acts via multiple mechanisms, we set out to pursue a targeted protein degradation strategy to abolish TG2's myriad functions. Here, we report the synthesis and characterization of a series of VHL-based degraders that reduce TG2 in ovarian cancer cells in a proteasome-dependent manner. Degradation of TG2 resulted in significantly reduced cancer cell adhesion and migration in vitro in scratch-wound and migration assays. These results strongly indicate that further development of more potent and in vivo efficient TG2 degraders could be a new strategy for reducing the dissemination of ovarian and other cancers.
Collapse
Affiliation(s)
- Andres Valdivia
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Purav P Vagadia
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Guangxu Guo
- WuXi AppTec, Shanghai 200131, People's Republic of China
| | - Eilidh O'Brien
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Daniela Matei
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Jesse Brown VA Medical Center, Chicago, Illinois 60612, United States
| | - Gary E Schiltz
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| |
Collapse
|
5
|
Byun DP, Ritchie J, Jung Y, Holewinski R, Kim HR, Tagirasa R, Ivanic J, Weekley CM, Parker MW, Andresson T, Yoo E. Covalent Inhibition by a Natural Product-Inspired Latent Electrophile. J Am Chem Soc 2023; 145:11097-11109. [PMID: 37183434 PMCID: PMC10719761 DOI: 10.1021/jacs.3c00598] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Strategies to target specific protein cysteines are critical to covalent probe and drug discovery. 3-Bromo-4,5-dihydroisoxazole (BDHI) is a natural product-inspired, synthetically accessible electrophilic moiety that has previously been shown to react with nucleophilic cysteines in the active site of purified enzymes. Here, we define the global cysteine reactivity and selectivity of a set of BDHI-functionalized chemical fragments using competitive chemoproteomic profiling methods. Our study demonstrates that BDHIs capably engage reactive cysteine residues in the human proteome and the selectivity landscape of cysteines liganded by BDHI is distinct from that of haloacetamide electrophiles. Given its tempered reactivity, BDHIs showed restricted, selective engagement with proteins driven by interactions between a tunable binding element and the complementary protein sites. We validate that BDHI forms covalent conjugates with glutathione S-transferase Pi (GSTP1) and peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (PIN1), emerging anticancer targets. BDHI electrophile was further exploited in Bruton's tyrosine kinase (BTK) inhibitor design using a single-step late-stage installation of the warhead onto acrylamide-containing compounds. Together, this study expands the spectrum of optimizable chemical tools for covalent ligand discovery and highlights the utility of 3-bromo-4,5-dihydroisoxazole as a cysteine-reactive electrophile.
Collapse
Affiliation(s)
- David P Byun
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Jennifer Ritchie
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Yejin Jung
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Ronald Holewinski
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biochemical Research, Frederick, Maryland 21702, United States
| | - Hong-Rae Kim
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Ravichandra Tagirasa
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Joseph Ivanic
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland 21702, United States
| | - Claire M Weekley
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Michael W Parker
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
- Australian Cancer Research Foundation Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Thorkell Andresson
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biochemical Research, Frederick, Maryland 21702, United States
| | - Euna Yoo
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| |
Collapse
|
6
|
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
|
7
|
Loppinet E, Besser HA, Sewa AS, Yang FC, Jabri B, Khosla C. LRP-1 links post-translational modifications to efficient presentation of celiac disease-specific T cell antigens. Cell Chem Biol 2023; 30:55-68.e10. [PMID: 36608691 PMCID: PMC9868102 DOI: 10.1016/j.chembiol.2022.12.002] [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: 07/25/2022] [Revised: 10/17/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023]
Abstract
Celiac disease (CeD) is an autoimmune disorder in which gluten-derived antigens trigger inflammation. Antigenic peptides must undergo site-specific deamidation to be presentable to CD4+ T cells in an HLA-DQ2 or -DQ8 restricted manner. While the biochemical basis for this post-translational modification is understood, its localization in the patient's intestine remains unknown. Here, we describe a mechanism by which gluten peptides undergo deamidation and concentration in the lysosomes of antigen-presenting cells, explaining how the concentration of gluten peptides necessary to elicit an inflammatory response in CeD patients is achieved. A ternary complex forms between a gluten peptide, transglutaminase-2 (TG2), and ubiquitous plasma protein α2-macroglobulin, and is endocytosed by LRP-1. The covalent TG2-peptide adduct undergoes endolysosomal decoupling, yielding the expected deamidated epitope. Our findings invoke a pathogenic role for dendritic cells and/or macrophages in CeD and implicate TG2 in the lysosomal clearance of unwanted self and foreign extracellular proteins.
Collapse
Affiliation(s)
- Elise Loppinet
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Harrison A Besser
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA; Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Agnele Sylvia Sewa
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Fu-Chen Yang
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Chaitan Khosla
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA; Department of Chemistry, Stanford University, Stanford, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
8
|
Lou J, Lu Y, Cheng J, Zhou F, Yan Z, Zhang D, Meng X, Zhao Y. A chemical perspective on the modulation of TEAD transcriptional activities: Recent progress, challenges, and opportunities. Eur J Med Chem 2022; 243:114684. [DOI: 10.1016/j.ejmech.2022.114684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/02/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022]
|
9
|
Tamanini E, Miyamura S, Buck IM, Cons BD, Dawson L, East C, Futamura T, Goto S, Griffiths-Jones C, Hashimoto T, Heightman TD, Ishikawa S, Ito H, Kaneko Y, Kawato T, Kondo K, Kurihara N, McCarthy JM, Mori Y, Nagase T, Nakaishi Y, Reeks J, Sato A, Schöpf P, Tai K, Tamai T, Tisi D, Woolford AJA. Fragment-Based Discovery of a Novel, Brain Penetrant, Orally Active HDAC2 Inhibitor. ACS Med Chem Lett 2022; 13:1591-1597. [PMID: 36262388 PMCID: PMC9575179 DOI: 10.1021/acsmedchemlett.2c00272] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022] Open
Abstract
Fragment-based ligand discovery was successfully applied to histone deacetylase HDAC2. In addition to the anticipated hydroxamic acid- and benzamide-based fragment screening hits, a low affinity (∼1 mM) α-amino-amide zinc binding fragment was identified, as well as fragments binding to other regions of the catalytic site. This alternative zinc-binding fragment was further optimized, guided by the structural information from protein-ligand complex X-ray structures, into a sub-μM, brain penetrant, HDAC2 inhibitor (17) capable of modulating histone acetylation levels in vivo.
Collapse
Affiliation(s)
- Emiliano Tamanini
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, U.K.
| | - Shin Miyamura
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Ildiko M. Buck
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, U.K.
| | - Benjamin D. Cons
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, U.K.
| | - Lee Dawson
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, U.K.
| | - Charlotte East
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, U.K.
| | - Takashi Futamura
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Shintaro Goto
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | | | - Tetsuya Hashimoto
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Tom D. Heightman
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, U.K.
| | - Shunpei Ishikawa
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Hideki Ito
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Yosuke Kaneko
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Tatsuya Kawato
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Kazumi Kondo
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Naoki Kurihara
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - James M. McCarthy
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, U.K.
| | - Yukiko Mori
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Tsuyoshi Nagase
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Yuichiro Nakaishi
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Judith Reeks
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, U.K.
| | - Akimasa Sato
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Patrick Schöpf
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, U.K.
| | - Kuninori Tai
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Taichi Tamai
- Otsuka
Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Dominic Tisi
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, U.K.
| | - Alison J.-A. Woolford
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, U.K.
| |
Collapse
|
10
|
Fadeeva AA, Ioffe SL, Tabolin AA. Synthesis of 3‐Haloisoxazolines by Deoxygenation of 3‐Haloisoxazoline
N
‐Oxides under Treatment with Acetyl Bromide. ChemistrySelect 2022. [DOI: 10.1002/slct.202202267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Anastasia A. Fadeeva
- N. D. Zelinsky Institute of Organic Chemistry Russian Academy of Sciences Leninsky prosp. 47 Moscow 119991 Russia
- Higher Chemical College D. Mendeleev University of Chemical Technology of Russia Miusskaya sq. 9 Moscow 125047 Russia
| | - Sema L. Ioffe
- N. D. Zelinsky Institute of Organic Chemistry Russian Academy of Sciences Leninsky prosp. 47 Moscow 119991 Russia
| | - Andrey A. Tabolin
- N. D. Zelinsky Institute of Organic Chemistry Russian Academy of Sciences Leninsky prosp. 47 Moscow 119991 Russia
| |
Collapse
|
11
|
Paolella G, Sposito S, Romanelli AM, Caputo I. Type 2 Transglutaminase in Coeliac Disease: A Key Player in Pathogenesis, Diagnosis and Therapy. Int J Mol Sci 2022; 23:ijms23147513. [PMID: 35886862 PMCID: PMC9318967 DOI: 10.3390/ijms23147513] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Type 2 transglutaminase (TG2) is the main autoantigen in coeliac disease (CD), a widespread inflammatory enteropathy caused by the ingestion of gluten-containing cereals in genetically predisposed individuals. As a consequence, serum antibodies to TG2 represent a very useful marker in CD diagnosis. However, TG2 is also an important player in CD pathogenesis, for its ability to deamidate some Gln residues of gluten peptides, which become more immunogenic in CD intestinal mucosa. Given the importance of TG2 enzymatic activities in CD, several studies have sought to discover specific and potent inhibitors that could be employed in new therapeutical approaches for CD, as alternatives to a lifelong gluten-free diet. In this review, we summarise all the aspects regarding TG2 involvement in CD, including its enzymatic reactions in pathogenesis, the role of anti-TG2 antibodies in disease management, and the exploration of recent strategies to reduce deamidation or to use transamidation to detoxify gluten.
Collapse
Affiliation(s)
- Gaetana Paolella
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano, SA, Italy; (S.S.); (A.M.R.)
- Correspondence: (G.P.); (I.C.)
| | - Silvia Sposito
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano, SA, Italy; (S.S.); (A.M.R.)
| | | | - Ivana Caputo
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano, SA, Italy; (S.S.); (A.M.R.)
- European Laboratory for the Investigation of Food-Induced Diseases (ELFID), University of Salerno, 84084 Fisciano, SA, Italy
- Correspondence: (G.P.); (I.C.)
| |
Collapse
|
12
|
He L, Hu X, Day DB, Yan M, Teng Y, Liu XL, Yan E, Xiang J, Qiu X, Mo J, Zhang Y, Zhang JJ, Gong J. The associations of nitrated polycyclic aromatic hydrocarbon exposures with plasma glucose and amino acids. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 289:117945. [PMID: 34426189 DOI: 10.1016/j.envpol.2021.117945] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/22/2021] [Accepted: 08/08/2021] [Indexed: 06/13/2023]
Abstract
Nitrated polycyclic aromatic hydrocarbons (nitro-PAHs) have been widely studied for their mutagenic and carcinogenic effects. This study aims to investigate whether exposure to nitro-PAHs is associated with biomarkers of carbohydrate metabolism, an underlying risk factor for metabolic disorder. Early morning urine and blood samples were longitudinally collected two times with a four-week interval from 43 healthy adults. Five urinary amino-PAHs (1-aminonaphthalene, 2-aminonaphthalene, 9-aminophenanthrene, 2-aminofluorene, and 1-aminopyrene) were measured as biomarkers of nitro-PAH exposures. We measured plasma concentrations of glucose and six amino acids that can regulate insulin secretion, including aspartate (Asp), glutamate (Glu), glutamine (Gln), alanine (Ala), Arginine (Arg), and ornithine (Orn). We found that increasing concentrations of 9-aminophenanthrene were significantly associated with increasing glucose levels and with decreasing Asp, Glu, Ala, and Orn levels. We estimated that 26.4 %-43.8 % of the 9-aminophenanthrene-associated increase in glucose level was mediated by Asp, Glu, and Orn. These results suggest that exposure to certain nitro-PAHs affects glucose homeostasis, partly resulting from the depletion of insulin-stimulating amino acids (Asp, Glu, and Orn).
Collapse
Affiliation(s)
- Linchen He
- Nicholas School of the Environment, Duke University, Durham, NC, 27708, USA; Global Health Institute, Duke University, Durham, NC, 27708, USA; Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Xinyan Hu
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China; Center for Environment and Health, Peking University, Beijing, 100871, China
| | - Drew B Day
- Seattle Children's Research Institute, Seattle, WA, 98145, United States
| | - Meilin Yan
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China; Center for Environment and Health, Peking University, Beijing, 100871, China
| | - Yanbo Teng
- Duke Kunshan University, Kunshan City, Jiangsu Province, 215316, China
| | - Xing Lucy Liu
- Global Health Institute, Duke University, Durham, NC, 27708, USA
| | - Erik Yan
- Global Health Institute, Duke University, Durham, NC, 27708, USA; Duke Kunshan University, Kunshan City, Jiangsu Province, 215316, China
| | - Jianbang Xiang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, United States
| | - Xinghua Qiu
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China; Center for Environment and Health, Peking University, Beijing, 100871, China
| | - Jinhan Mo
- Department of Building Science, Tsinghua University, Beijing, 100084, China; Beijing Key Laboratory of Indoor Air Quality Evaluation and Control, Beijing, 100084, China
| | - Yinping Zhang
- Department of Building Science, Tsinghua University, Beijing, 100084, China; Beijing Key Laboratory of Indoor Air Quality Evaluation and Control, Beijing, 100084, China
| | - Junfeng Jim Zhang
- Nicholas School of the Environment, Duke University, Durham, NC, 27708, USA; Global Health Institute, Duke University, Durham, NC, 27708, USA; Duke Kunshan University, Kunshan City, Jiangsu Province, 215316, China
| | - Jicheng Gong
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China; Center for Environment and Health, Peking University, Beijing, 100871, China.
| |
Collapse
|
13
|
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: 13] [Impact Index Per Article: 3.3] [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
|
14
|
Zana A, Galbiati A. Synthesis and Reactivity of 3‐Halo‐4,5‐dihydroisoxazoles: An Overview. ChemistrySelect 2021. [DOI: 10.1002/slct.202101779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Aureliano Zana
- Department of Pharmaceutical Sciences Università degli Studi di Milano Via Mangiagalli 25 20133 Milano Italy
- Philochem AG Libernstrasse 3 8112 Otelfingen (ZH) Switzerland
| | - Andrea Galbiati
- Department of Pharmaceutical Sciences Università degli Studi di Milano Via Mangiagalli 25 20133 Milano Italy
- Philochem AG Libernstrasse 3 8112 Otelfingen (ZH) Switzerland
| |
Collapse
|
15
|
Probing tissue transglutaminase mediated vascular smooth muscle cell aging using a novel transamidation-deficient Tgm2-C277S mouse model. Cell Death Discov 2021; 7:197. [PMID: 34326316 PMCID: PMC8322091 DOI: 10.1038/s41420-021-00543-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/21/2021] [Accepted: 05/29/2021] [Indexed: 02/07/2023] Open
Abstract
Tissue transglutaminase (TG2), a multifunctional protein of the transglutaminase family, has putative transamidation-independent functions in aging-associated vascular stiffening and dysfunction. Developing preclinical models will be critical to fully understand the physiologic relevance of TG2's transamidation-independent activity and to identify the specific function of TG2 for therapeutic targeting. Therefore, in this study, we harnessed CRISPR-Cas9 gene editing technology to introduce a mutation at cysteine 277 in the active site of the mouse Tgm2 gene. Heterozygous and homozygous Tgm2-C277S mice were phenotypically normal and were born at the expected Mendelian frequency. TG2 protein was ubiquitously expressed in the Tgm2-C277S mice at levels similar to those of wild-type (WT) mice. In the Tgm2-C277S mice, TG2 transglutaminase function was successfully obliterated, but the transamidation-independent functions ascribed to GTP, fibronectin, and integrin binding were preserved. In vitro, a remodeling stimulus led to the significant loss of vascular compliance in WT mice, but not in the Tgm2-C277S or TG2-/- mice. Vascular stiffness increased with age in WT mice, as measured by pulse-wave velocity and tensile testing. Tgm2-C277S mice were protected from age-associated vascular stiffening, and TG2 knockout yielded further protection. Together, these studies show that TG2 contributes significantly to overall vascular modulus and vasoreactivity independent of its transamidation function, but that transamidation activity is a significant cause of vascular matrix stiffening during aging. Finally, the Tgm2-C277S mice can be used for in vivo studies to explore the transamidation-independent roles of TG2 in physiology and pathophysiology.
Collapse
|
16
|
|
17
|
Prat-Duran J, Pinilla E, Nørregaard R, Simonsen U, Buus NH. Transglutaminase 2 as a novel target in chronic kidney disease - Methods, mechanisms and pharmacological inhibition. Pharmacol Ther 2020; 222:107787. [PMID: 33307141 DOI: 10.1016/j.pharmthera.2020.107787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/02/2020] [Indexed: 01/31/2023]
Abstract
Chronic kidney disease (CKD) is a global health problem with a prevalence of 10-15%. Progressive fibrosis of the renal tissue is a main feature of CKD, but current treatment strategies are relatively unspecific and delay, but do not prevent, CKD. Exploration of novel pharmacological targets to inhibit fibrosis development are therefore important. Transglutaminase 2 (TG2) is known to be central for extracellular collagenous matrix formation, but TG2 is a multifunctional enzyme and novel research has broadened our view on its extra- and intracellular actions. TG2 exists in two conformational states with different catalytic properties as determined by substrate availability and local calcium concentrations. The open conformation of TG2 depends on calcium and has transamidase activity, central for protein modification and cross-linking of extracellular protein components, while the closed conformation is a GTPase involved in transmembrane signaling processes. We first describe different methodologies to assess TG2 activity in renal tissue and cell cultures such as biotin cadaverine incorporation. Then we systematically review animal CKD models and preliminary studies in humans (with diabetic, IgA- and chronic allograft nephropathy) to reveal the role of TG2 in renal fibrosis. Mechanisms behind TG2 activation, TG2 externalization dependent on Syndecan-4 and interactions between TG and profibrotic molecules including transforming growth factor β and the angiotensin II receptor are discussed. Pharmacological TG2 inhibition shows antifibrotic effects in CKD. However, the translation of TG2 inhibition to treat CKD in patients is a challenge as clinical information is limited, and further studies on pharmacokinetics and efficacy of the individual compounds are required.
Collapse
Affiliation(s)
| | | | | | - Ulf Simonsen
- Institute of Biomedicine, Health, Aarhus University, Denmark
| | - Niels Henrik Buus
- Institute of Biomedicine, Health, Aarhus University, Denmark; Department of Renal Medicine, Aarhus University Hospital, Denmark.
| |
Collapse
|
18
|
Galbiati A, Zana A, Conti P. Covalent inhibitors of GAPDH: From unspecific warheads to selective compounds. Eur J Med Chem 2020; 207:112740. [PMID: 32898762 DOI: 10.1016/j.ejmech.2020.112740] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/23/2020] [Accepted: 08/05/2020] [Indexed: 11/18/2022]
Abstract
Targeting glycolysis is an attractive approach for the treatment of a wide range of pathologies, such as various tumors and parasitic infections. Due to its pivotal role in the glycolysis, Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) represents a rate-limiting enzyme in those cells that mostly, or exclusively rely on this pathway for energy production. In this context, GAPDH inhibition can be a valuable approach for the development of anticancer and antiparasitic drugs. In addition to its glycolytic role, GAPDH possesses several moonlight functions, whose deregulation is involved in some pathological conditions. Covalent modification on different amino acids of GAPDH, in particular on cysteine residues, can lead to a modulation of the enzyme activity. The selectivity towards specific cysteine residues is essential to achieve a specific phenotypic effect. In this work we report an extensive overview of the latest advances on the numerous compounds able to inhibit GAPDH through the covalent binding to cysteine residues, ranging from endogenous metabolites and xenobiotics, which may serve as pharmacological tools to actual drug-like compounds with promising therapeutic perspectives. Furthermore, we focused on the potentialities of the different warheads, shedding light on the possibility to exploit a combination of a finely tuned electrophilic group with a well-designed recognition moiety. These findings can provide useful information for the rational design of novel covalent inhibitors of GAPDH, with the final goal to expand the current treatment options.
Collapse
Affiliation(s)
- Andrea Galbiati
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via Mangiagalli 25, 20133, Milano, Italy.
| | - Aureliano Zana
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via Mangiagalli 25, 20133, Milano, Italy
| | - Paola Conti
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via Mangiagalli 25, 20133, Milano, Italy
| |
Collapse
|
19
|
Martucciello S, Sposito S, Esposito C, Paolella G, Caputo I. Interplay between Type 2 Transglutaminase (TG2), Gliadin Peptide 31-43 and Anti-TG2 Antibodies in Celiac Disease. Int J Mol Sci 2020; 21:ijms21103673. [PMID: 32456177 PMCID: PMC7279455 DOI: 10.3390/ijms21103673] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 02/07/2023] Open
Abstract
Celiac disease (CD) is a common intestinal inflammatory disease involving both a genetic background and environmental triggers. The ingestion of gluten, a proteic component of several cereals, represents the main hexogen factor implied in CD onset that involves concomitant innate and adaptive immune responses to gluten. Immunogenicity of some gluten sequences are strongly enhanced as the consequence of the deamidation of specific glutamine residues by type 2 transglutaminase (TG2), a ubiquitous enzyme whose expression is up-regulated in the intestine of CD patients. A short gluten sequence resistant to intestinal proteases, the α-gliadin peptide 31-43, seems to modulate TG2 function in the gut; on the other hand, the enzyme can affect the biological activity of this peptide. In addition, an intense auto-immune response towards TG2 is a hallmark of CD. Auto-antibodies exert a range of biological effects on several cells, effects that in part overlap with those induced by peptide 31-43. In this review, we delineate a scenario in which TG2, anti-TG2 antibodies and peptide 31-43 closely relate to each other, thus synergistically participating in CD starting and progression.
Collapse
Affiliation(s)
- Stefania Martucciello
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano (SA), Italy; (S.M.); (C.E.); (G.P.)
| | - Silvia Sposito
- European Laboratory for the Investigation of Food-Induced Diseases (ELFID), University of Salerno, 84084 Fisciano (SA), Italy;
| | - Carla Esposito
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano (SA), Italy; (S.M.); (C.E.); (G.P.)
- European Laboratory for the Investigation of Food-Induced Diseases (ELFID), University of Salerno, 84084 Fisciano (SA), Italy;
| | - Gaetana Paolella
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano (SA), Italy; (S.M.); (C.E.); (G.P.)
| | - Ivana Caputo
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano (SA), Italy; (S.M.); (C.E.); (G.P.)
- European Laboratory for the Investigation of Food-Induced Diseases (ELFID), University of Salerno, 84084 Fisciano (SA), Italy;
- Correspondence: ; Tel.: +39-089-969592; Fax: +39-089-969603
| |
Collapse
|
20
|
Abadie V, Kim SM, Lejeune T, Palanski BA, Ernest JD, Tastet O, Voisine J, Discepolo V, Marietta EV, Hawash MBF, Ciszewski C, Bouziat R, Panigrahi K, Horwath I, Zurenski MA, Lawrence I, Dumaine A, Yotova V, Grenier JC, Murray JA, Khosla C, Barreiro LB, Jabri B. IL-15, gluten and HLA-DQ8 drive tissue destruction in coeliac disease. Nature 2020; 578:600-604. [PMID: 32051586 PMCID: PMC7047598 DOI: 10.1038/s41586-020-2003-8] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 12/16/2019] [Indexed: 11/09/2022]
Abstract
Coeliac disease is a complex, polygenic inflammatory enteropathy caused by exposure to dietary gluten that occurs in a subset of genetically susceptible individuals who express either the HLA-DQ8 or HLA-DQ2 haplotypes1,2. The need to develop non-dietary treatments is now widely recognized3, but no pathophysiologically relevant gluten- and HLA-dependent preclinical model exists. Furthermore, although studies in humans have led to major advances in our understanding of the pathogenesis of coeliac disease4, the respective roles of disease-predisposing HLA molecules, and of adaptive and innate immunity in the development of tissue damage, have not been directly demonstrated. Here we describe a mouse model that reproduces the overexpression of interleukin-15 (IL-15) in the gut epithelium and lamina propria that is characteristic of active coeliac disease, expresses the predisposing HLA-DQ8 molecule, and develops villous atrophy after ingestion of gluten. Overexpression of IL-15 in both the epithelium and the lamina propria is required for the development of villous atrophy, which demonstrates the location-dependent central role of IL-15 in the pathogenesis of coeliac disease. In addition, CD4+ T cells and HLA-DQ8 have a crucial role in the licensing of cytotoxic T cells to mediate intestinal epithelial cell lysis. We also demonstrate a role for the cytokine interferon-γ (IFNγ) and the enzyme transglutaminase 2 (TG2) in tissue destruction. By reflecting the complex interaction between gluten, genetics and IL-15-driven tissue inflammation, this mouse model provides the opportunity to both increase our understanding of coeliac disease, and develop new therapeutic strategies.
Collapse
Affiliation(s)
- Valérie Abadie
- Department of Microbiology, Infectiology, and Immunology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada.
- Sainte-Justine Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada.
- Department of Medicine, University of Chicago, Chicago, IL, USA.
| | - Sangman M Kim
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Department of Biology, University of San Francisco, San Francisco, CA, USA
| | - Thomas Lejeune
- Department of Microbiology, Infectiology, and Immunology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Sainte-Justine Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Brad A Palanski
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Jordan D Ernest
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Olivier Tastet
- Department of Genetics, Sainte-Justine Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Jordan Voisine
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | | | - Eric V Marietta
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Dermatology, Mayo Clinic, Rochester, MN, USA
| | - Mohamed B F Hawash
- Department of Genetics, Sainte-Justine Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
- Department of Biochemistry, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Cezary Ciszewski
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Romain Bouziat
- Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | | | - Irina Horwath
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | - Ian Lawrence
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Anne Dumaine
- Department of Genetics, Sainte-Justine Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Vania Yotova
- Department of Genetics, Sainte-Justine Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Jean-Christophe Grenier
- Department of Genetics, Sainte-Justine Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Joseph A Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Chaitan Khosla
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Stanford ChEM-H, Stanford University, Stanford, CA, USA
| | - Luis B Barreiro
- Department of Genetics, Sainte-Justine Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
- Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, IL, USA
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, IL, USA.
- Committee on Immunology, University of Chicago, Chicago, IL, USA.
- Department of Pediatrics, Section of Gastroenterology, Hepatology and Nutrition, University of Chicago, Chicago, IL, USA.
- Department of Pathology, University of Chicago, Chicago, IL, USA.
- University of Chicago Celiac Disease Center, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
21
|
Transglutaminases, neuronal cell death and neural repair: implications for traumatic brain injury and therapeutics. Curr Opin Neurol 2019; 32:796-801. [DOI: 10.1097/wco.0000000000000753] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
22
|
Zhang J, Wang L, Xiang J, Cui J, Hu B, Yang L, Tang Y. HOAc‐Assisted Synthesis of 2,3‐Disubstituted Quinolines from Arylamine and Aliphatic Aldehyde in Water. ChemistrySelect 2019. [DOI: 10.1002/slct.201901686] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Jing‐Tao Zhang
- Key Laboratory for Environmentally Friendly Chemistry and ApplicationDepartment of ChemistryXiangtan University Hunan 411105 P. R. China
- Beijing National Laboratory for Molecular Sciences (BNLMS)Center for Molecular Sciences, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of ChemistryChinese Academy of Sciences Beijing 100190 P. R. China
| | - Li–Xia Wang
- Beijing National Laboratory for Molecular Sciences (BNLMS)Center for Molecular Sciences, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of ChemistryChinese Academy of Sciences Beijing 100190 P. R. China
| | - Jun‐Feng Xiang
- Beijing National Laboratory for Molecular Sciences (BNLMS)Center for Molecular Sciences, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of ChemistryChinese Academy of Sciences Beijing 100190 P. R. China
| | - Jie Cui
- Beijing National Laboratory for Molecular Sciences (BNLMS)Center for Molecular Sciences, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of ChemistryChinese Academy of Sciences Beijing 100190 P. R. China
| | - Ben‐Quan Hu
- Key Laboratory for Environmentally Friendly Chemistry and ApplicationDepartment of ChemistryXiangtan University Hunan 411105 P. R. China
| | - Luo Yang
- Key Laboratory for Environmentally Friendly Chemistry and ApplicationDepartment of ChemistryXiangtan University Hunan 411105 P. R. China
| | - Ya‐Lin Tang
- Beijing National Laboratory for Molecular Sciences (BNLMS)Center for Molecular Sciences, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of ChemistryChinese Academy of Sciences Beijing 100190 P. R. China
- University of the Chinese Academy of Sciences Beijing 100049 PR China
| |
Collapse
|
23
|
Rao MS, Sarkar S, Hussain S. Microwave-assisted synthesis of 3-aminoarylquinolines from 2-nitrobenzaldehyde and indole via SnCl2-mediated reduction and facile indole ring opening. Tetrahedron Lett 2019. [DOI: 10.1016/j.tetlet.2019.03.047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
24
|
Takamura H, Tanaka M, Ando J, Tazawa A, Ishizawa K. Stereodivergent and Stereoselective Synthesis of cis- and trans-4-Substituted Prolinols. HETEROCYCLES 2019. [DOI: 10.3987/com-18-s(f)8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
Takamura H, Tanaka M, Ando J, Tazawa A, Ishizawa K. Concise Synthesis of Anticancer Active trans-4-(4-Octylphenyl)prolinol. HETEROCYCLES 2019. [DOI: 10.3987/com-18-s(f)44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
26
|
Gao Y, Hider RC, Ma Y. An efficient 3-acylquinoline synthesis from acetophenones and anthranilviaC(sp3)–H bond activation mediated by Selectfluor. RSC Adv 2019; 9:10340-10344. [PMID: 35520934 PMCID: PMC9062302 DOI: 10.1039/c9ra01481k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 03/27/2019] [Indexed: 01/01/2023] Open
Abstract
An efficient method for the synthesis of 3-functionalized quinolines from commercially available acetophenones and anthranil has been described. Selectfluor propels the C(sp3)–H bond activation of the acetophenones and aza-Michael addition of anthranil resulting in annulated 3-acylquinolines in moderate to high yields. DMSO acts not only as a solvent but also as a one carbon donor in the reaction. An efficient method for the synthesis of 3-functionalized quinolines from acetophenones and anthranil mediated by Selectfluor was achieved.![]()
Collapse
Affiliation(s)
- Yejun Gao
- School of Pharmaceutical and Chemical Engineering
- Taizhou University
- Taizhou 318000
- PR China
- School of Pharmaceutical Science
| | - Robert C. Hider
- Institute of Pharmaceutical Science
- King's College London
- London
- UK
| | - Yongmin Ma
- School of Pharmaceutical and Chemical Engineering
- Taizhou University
- Taizhou 318000
- PR China
- School of Pharmaceutical Science
| |
Collapse
|
27
|
Cystamine and cysteamine as inhibitors of transglutaminase activity in vivo. Biosci Rep 2018; 38:BSR20180691. [PMID: 30054429 PMCID: PMC6123069 DOI: 10.1042/bsr20180691] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 11/07/2018] [Accepted: 07/24/2018] [Indexed: 12/22/2022] Open
Abstract
Cystamine is commonly used as a transglutaminase inhibitor. This disulphide undergoes reduction in vivo to the aminothiol compound, cysteamine. Thus, the mechanism by which cystamine inhibits transglutaminase activity in vivo could be due to either cystamine or cysteamine, which depends on the local redox environment. Cystamine inactivates transglutaminases by promoting the oxidation of two vicinal cysteine residues on the enzyme to an allosteric disulphide, whereas cysteamine acts as a competitive inhibitor for transamidation reactions catalyzed by this enzyme. The latter mechanism is likely to result in the formation of a unique biomarker, N-(γ-glutamyl)cysteamine that could serve to indicate how cyst(e)amine acts to inhibit transglutaminases inside cells and the body.
Collapse
|
28
|
Phanindrudu M, Wakade SB, Tiwari DK, Likhar PR, Tiwari DK. Transition-Metal-Free Approach for the Synthesis of 4-Aryl-quinolines from Alkynes and Anilines. J Org Chem 2018; 83:9137-9143. [DOI: 10.1021/acs.joc.8b01204] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Mandalaparthi Phanindrudu
- Division of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi 110001, India
| | - Sandip Balasaheb Wakade
- Division of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi 110001, India
| | - Dipak Kumar Tiwari
- Division of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi 110001, India
| | - Pravin R. Likhar
- Inorganic and Physical Chemistry Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi 110001, India
| | - Dharmendra Kumar Tiwari
- Division of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi 110001, India
| |
Collapse
|
29
|
Cardiac fibrosis can be attenuated by blocking the activity of transglutaminase 2 using a selective small-molecule inhibitor. Cell Death Dis 2018; 9:613. [PMID: 29795262 PMCID: PMC5966415 DOI: 10.1038/s41419-018-0573-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 04/01/2018] [Accepted: 04/03/2018] [Indexed: 12/31/2022]
Abstract
Cardiac fibrosis is implicit in all forms of heart disease but there are no effective treatments. In this report, we investigate the role of the multi-functional enzyme Transglutaminase 2 (TG2) in cardiac fibrosis and assess its potential as a therapeutic target. Here we describe the use a highly selective TG2 small-molecule inhibitor to test the efficacy of TG2 inhibition as an anti-fibrotic therapy for heart failure employing two different in vivo models of cardiac fibrosis: Progressively induced interstitial cardiac fibrosis by pressure overload using angiotensin II infusion: Acutely induced focal cardiac fibrosis through myocardial infarction by ligation of the left anterior descending coronary artery (AMI model). In the AMI model, in vivo MRI showed that the TG2 inhibitor 1–155 significantly reduced infarct size by over 50% and reduced post-infarct remodelling at 20 days post insult. In both models, Sirius red staining for collagen deposition and levels of the TG2-mediated protein crosslink ε(γ-glutamyl)lysine were significantly reduced. No cardiac rupture or obvious signs of toxicity were observed. To provide a molecular mechanism for TG2 involvement in cardiac fibrosis, we show that both TGFβ1-induced transition of cardiofibroblasts into myofibroblast-like cells and TGFβ1-induced EndMT, together with matrix deposition, can be attenuated by the TG2 selective inhibitor 1–155, suggesting a new role for TG2 in regulating TGFβ1 signalling in addition to its role in latent TGFβ1 activation. In conclusion, TG2 has a role in cardiac fibrosis through activation of myofibroblasts and matrix deposition. TG2 inhibition using a selective small-molecule inhibitor can attenuate cardiac fibrosis.
Collapse
|
30
|
Wodtke R, Hauser C, Ruiz-Gómez G, Jäckel E, Bauer D, Lohse M, Wong A, Pufe J, Ludwig FA, Fischer S, Hauser S, Greif D, Pisabarro MT, Pietzsch J, Pietsch M, Löser R. Nε-Acryloyllysine Piperazides as Irreversible Inhibitors of Transglutaminase 2: Synthesis, Structure–Activity Relationships, and Pharmacokinetic Profiling. J Med Chem 2018; 61:4528-4560. [DOI: 10.1021/acs.jmedchem.8b00286] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Robert Wodtke
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
- Fakultät Natur- und Umweltwissenschaften, Hochschule Zittau/Görlitz, Theodor-Körner-Allee 16, 02763 Zittau, Germany
- Fakultät Chemie und Lebensmittelchemie, Technische Universität Dresden, Mommsenstraße 4, 01062 Dresden, Germany
| | - Christoph Hauser
- Zentrum für Pharmakologie, Medizinische Fakultät, Universität zu Köln, Gleueler Straße 24, 50931 Köln, Germany
| | - Gloria Ruiz-Gómez
- Structural Bioinformatics, BIOTEC, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Elisabeth Jäckel
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
- Fakultät Natur- und Umweltwissenschaften, Hochschule Zittau/Görlitz, Theodor-Körner-Allee 16, 02763 Zittau, 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
| | - Martin Lohse
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
- Fakultät Natur- und Umweltwissenschaften, Hochschule Zittau/Görlitz, Theodor-Körner-Allee 16, 02763 Zittau, Germany
| | - Alan Wong
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Johanna Pufe
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Friedrich-Alexander Ludwig
- Institut für Radiopharmazeutische Krebsforschung, Forschungsstelle Leipzig, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße 15, 04318 Leipzig, Germany
| | - Steffen Fischer
- Institut für Radiopharmazeutische Krebsforschung, Forschungsstelle Leipzig, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße 15, 04318 Leipzig, Germany
| | - Sandra Hauser
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Dieter Greif
- Fakultät Natur- und Umweltwissenschaften, Hochschule Zittau/Görlitz, Theodor-Körner-Allee 16, 02763 Zittau, Germany
| | - M. Teresa Pisabarro
- Structural Bioinformatics, BIOTEC, Technische Universität Dresden, Tatzberg 47-51, 01307 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
| | - Markus Pietsch
- Zentrum für Pharmakologie, Medizinische Fakultät, Universität zu Köln, Gleueler Straße 24, 50931 Köln, 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
|
31
|
Palanski BA, Khosla C. Cystamine and Disulfiram Inhibit Human Transglutaminase 2 via an Oxidative Mechanism. Biochemistry 2018; 57:3359-3363. [PMID: 29570977 DOI: 10.1021/acs.biochem.8b00204] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The catalytic activity of transglutaminase 2 (TG2), a ubiquitously expressed mammalian enzyme, is regulated by multiple post-translational mechanisms. Because elevated activity of TG2 in the extracellular matrix is associated with organ-specific diseases such as celiac disease and renal fibrosis, there is growing therapeutic interest in inhibitors of this enzyme. Cystamine, a symmetric disulfide compound, is one of the earliest reported TG2 inhibitors. Despite its widespread use as a tool compound to block TG2 activity in vitro and in vivo, its mechanism of action has remained unclear. Here, we demonstrate that cystamine irreversibly inhibits human TG2 ( kinh/ Ki = 1.2 mM-1 min-1) via a mechanism fundamentally distinct from those proposed previously. Through mass spectrometric disulfide mapping and site-directed mutagenesis, we show that cystamine promotes the formation of a physiologically relevant disulfide bond between Cys370 and Cys371 that allosterically abrogates the catalytic activity of human TG2. This discovery led us to evaluate clinically useful thiol → disulfide oxidants for TG2 inhibitory activity. It is demonstrated that disulfiram, a relatively safe oral thiuram disulfide, is a fairly potent TG2 inhibitor ( kinh/ Ki = 8.3 mM-1 min-1) and may therefore provide a practical tool for clinically validating this emerging therapeutic target in intestinal disorders such as celiac disease.
Collapse
|
32
|
Yu LZ, Wei HZ, Shi M. Base-Catalyzed Cascade Reaction of ortho
-(Propargylamino)aryl Ketones with N-, O-, or S-Based Nucleophiles for the Synthesis of 3-Functionalized Quinoline Scaffolds. Adv Synth Catal 2018. [DOI: 10.1002/adsc.201800120] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Liu-Zhu Yu
- State Key Laboratory of Organometallic Chemistry; Center for Excellence in Molecular Synthesis; Shanghai Institute of Organic Chemistry; University of Chinese Academy of Science; Chinese Academy of Sciences; 345 Lingling Road Shanghai 200032 People's Republic of China
| | - Hao-Zhao Wei
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals; School of Chemistry & Molecular Engineering; East China University of Science and Technology; 130 Mei Long Road Shanghai 200237 People's Republic of China
| | - Min Shi
- State Key Laboratory of Organometallic Chemistry; Center for Excellence in Molecular Synthesis; Shanghai Institute of Organic Chemistry; University of Chinese Academy of Science; Chinese Academy of Sciences; 345 Lingling Road Shanghai 200032 People's Republic of China
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals; School of Chemistry & Molecular Engineering; East China University of Science and Technology; 130 Mei Long Road Shanghai 200237 People's Republic of China
- State Key Laboratory and Institute of Elemento-organic Chemistry; Nankai University; Tianjin 300071 People's Republic of China
| |
Collapse
|
33
|
Valenti S, Corica D, Ricciardi L, Romano C. Gluten-related disorders: certainties, questions and doubts. Ann Med 2017; 49:569-581. [PMID: 28462603 DOI: 10.1080/07853890.2017.1325968] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/24/2017] [Indexed: 02/07/2023] Open
Abstract
In the last decade, the ingestion of gluten, a heterogeneous complex of proteins present in wheat, rice, barley and probably in oats, has been associated with clinical disorders, such as celiac disease, wheat allergy and recently to non-celiac gluten sensitivity or wheat intolerance syndrome. Gluten-related disorders, which are becoming epidemiologically relevant with an estimated global prevalence of about 5%, require the exclusion of gluten from the diet. For the past 5 years, an important shift in the availability of gluten-free products, together with increased consumption in the general population, has been recorded and is estimated to be about 12-25%. Many people follow a self-prescribed gluten-free diet, despite the fact that the majority have not first been previously excluded, or confirmed, as having gluten disorders. They rely on claims that a gluten-free diet improves general health. In this review, we provide an overview of the clinical disorders related to gluten or wheat ingestion, pointing out the current certainties, open questions, possible answers and several doubts in the management of these conditions. KEY MESSAGE Incidence of gluten-related disorders is increased in the last decade and self-diagnosis is frequent with inappropriate starting of a gluten-free diet. Gluten and wheat are considered as the most important triggers to coeliac disease, wheat allergy and non-celiac gluten sensitivity. Pediatricians, allergologist and gastroenterologist are involved in the management of these conditions and appropriate diagnostic protocols are required.
Collapse
Affiliation(s)
- Simona Valenti
- a Pediatrics Unit, Department of Human Pathology in Adulthood and Childhood "G. Barresi" , University of Messina , Messina , Italy
| | - Domenico Corica
- a Pediatrics Unit, Department of Human Pathology in Adulthood and Childhood "G. Barresi" , University of Messina , Messina , Italy
| | - Luisa Ricciardi
- b Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine , University of Messina , Messina , Italy
| | - Claudio Romano
- a Pediatrics Unit, Department of Human Pathology in Adulthood and Childhood "G. Barresi" , University of Messina , Messina , Italy
| |
Collapse
|
34
|
Wakade SB, Tiwari DK, Ganesh PSKP, Phanindrudu M, Likhar PR, Tiwari DK. Transition-Metal-Free Quinoline Synthesis from Acetophenones and Anthranils via Sequential One-Carbon Homologation/Conjugate Addition/Annulation Cascade. Org Lett 2017; 19:4948-4951. [DOI: 10.1021/acs.orglett.7b02429] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Sandip Balasaheb Wakade
- Medicinal
Chemistry and Biotechnology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi 110001, India
| | - Dipak Kumar Tiwari
- Medicinal
Chemistry and Biotechnology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi 110001, India
| | | | - Mandalaparthi Phanindrudu
- Medicinal
Chemistry and Biotechnology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi 110001, India
| | - Pravin R. Likhar
- Academy of Scientific & Innovative Research (AcSIR), New Delhi 110001, India
| | - Dharmendra Kumar Tiwari
- Medicinal
Chemistry and Biotechnology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi 110001, India
| |
Collapse
|
35
|
Szondy Z, Korponay-Szabó I, Király R, Sarang Z, Tsay GJ. Transglutaminase 2 in human diseases. Biomedicine (Taipei) 2017; 7:15. [PMID: 28840829 PMCID: PMC5571667 DOI: 10.1051/bmdcn/2017070315] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 05/15/2017] [Indexed: 12/30/2022] Open
Abstract
Transglutaminase 2 (TG2) is an inducible transamidating acyltransferase that catalyzes Ca(2+)-dependent protein modifications. In addition to being an enzyme, TG2 also serves as a G protein for several seven transmembrane receptors and acts as a co-receptor for integrin β1 and β3 integrins distinguishing it from other members of the transglutaminase family. TG2 is ubiquitously expressed in almost all cell types and all cell compartments, and is also present on the cell surface and gets secreted to the extracellular matrix via non-classical mechanisms. TG2 has been associated with various human diseases including inflammation, cancer, fibrosis, cardiovascular disease, neurodegenerative diseases, celiac disease in which it plays either a protective role, or contributes to the pathogenesis. Thus modulating the biological activities of TG2 in these diseases will have a therapeutic value.
Collapse
Affiliation(s)
- Zsuzsa Szondy
- Dental Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen 4010, Hungary
| | - Ilma Korponay-Szabó
- Department of Pediatrics and Biochemistry and Molecular Biology, University of Debrecen, Debrecen 4010, Hungary - Celiac Disease Center, Heim Pál Children's Hospital, Budapest 1089, Hungary
| | - Robert Király
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen 4010, Hungary
| | - Zsolt Sarang
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen 4010, Hungary
| | - Gregory J Tsay
- Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan - School of medicine, College of Medicine, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
36
|
Plugis NM, Palanski BA, Weng CH, Albertelli M, Khosla C. Thioredoxin-1 Selectively Activates Transglutaminase 2 in the Extracellular Matrix of the Small Intestine: IMPLICATIONS FOR CELIAC DISEASE. J Biol Chem 2016; 292:2000-2008. [PMID: 28003361 DOI: 10.1074/jbc.m116.767988] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/13/2016] [Indexed: 11/06/2022] Open
Abstract
Transglutaminase 2 (TG2) catalyzes transamidation or deamidation of its substrates and is ordinarily maintained in a catalytically inactive state in the intestine and other organs. Aberrant TG2 activity is thought to play a role in celiac disease, suggesting that a better understanding of TG2 regulation could help to elucidate the mechanistic basis of this malady. Structural and biochemical analysis has led to the hypothesis that extracellular TG2 activation involves reduction of an allosteric disulfide bond by thioredoxin-1 (TRX), but cellular and in vivo evidence for this proposal is lacking. To test the physiological relevance of this hypothesis, we first showed that macrophages exposed to pro-inflammatory stimuli released TRX in sufficient quantities to activate their extracellular pools of TG2. By using the C35S mutant of TRX, which formed a metastable mixed disulfide bond with TG2, we demonstrated that these proteins specifically recognized each other in the extracellular matrix of fibroblasts. When injected into mice and visualized with antibodies, we observed the C35S TRX mutant bound to endogenous TG2 as its principal protein partner in the small intestine. Control experiments showed no labeling of TG2 knock-out mice. Intravenous administration of recombinant TRX in wild-type mice, but not TG2 knock-out mice, led to a rapid rise in intestinal transglutaminase activity in a manner that could be inhibited by small molecules targeting TG2 or TRX. Our findings support the potential pathophysiological relevance of TRX in celiac disease and establish the Cys370-Cys371 disulfide bond of TG2 as one of clearest examples of an allosteric disulfide bond in mammals.
Collapse
Affiliation(s)
- Nicholas M Plugis
- From the Department of Chemistry, Stanford University, Stanford, California 94305
| | - Brad A Palanski
- From the Department of Chemistry, Stanford University, Stanford, California 94305
| | - Chih-Hisang Weng
- From the Department of Chemistry, Stanford University, Stanford, California 94305; the School of Medicine, Stanford University, Stanford, California 94305; the Medical Science Training Program, Stanford University, Stanford, California 94305
| | - Megan Albertelli
- Department of Comparative Medicine, Stanford University, Stanford, California 94305
| | - Chaitan Khosla
- From the Department of Chemistry, Stanford University, Stanford, California 94305; Department of Chemical Engineering, Stanford University, Stanford, California 94305; Stanford ChEM-H, Stanford University, Stanford, California 94305.
| |
Collapse
|
37
|
Song M, Hwang H, Im CY, Kim SY. Recent Progress in the Development of Transglutaminase 2 (TGase2) Inhibitors. J Med Chem 2016; 60:554-567. [PMID: 28122456 DOI: 10.1021/acs.jmedchem.6b01036] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Transglutaminase 2 (TGase2, TG2) activity has been implicated in the pathogenesis of a number of unrelated disorders, including celiac, neurological, and renal diseases, and various forms of cancer. It has been suggested that TGase2 activity, such as cross-linking, deamidation, and GTP-related activity, is associated with each disease. Continuing efforts to develop small molecule TG2 inhibitors are ongoing. To develop a new class of TG2 inhibitors, the factors impeding the development of TG2 inhibitors have been identified. Additionally, the conformational effect of TG2 enzyme in regard to its pathological roles, in vitro screening methods, recently discovered TG2 inhibitors, and preclinical evaluations are discussed with a brief summary of current TG2 inhibitor pipelines under the clinical setting.
Collapse
Affiliation(s)
- Minsoo Song
- New Drug Development Center (NDDC), Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF) , 80 Cheombok-ro, Dong-gu, Daegu 701-310, Korea
| | - Hayoung Hwang
- New Drug Development Center (NDDC), Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF) , 80 Cheombok-ro, Dong-gu, Daegu 701-310, Korea
| | - Chun Young Im
- New Drug Development Center (NDDC), Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF) , 80 Cheombok-ro, Dong-gu, Daegu 701-310, Korea
| | - Soo-Youl Kim
- Cancer Cell and Molecular Biology Branch, Research Institute, National Cancer Center , Goyang-si, Gyeonggi-do 410-769, Korea
| |
Collapse
|
38
|
Guo S, Palanski BA, Kloeck C, Khosla C, Cui B. Intracellular TG2 Activity Increases Microtubule Stability but is not Sufficient to Prompt Neurite Growth. Neurosci Bull 2016; 33:103-106. [PMID: 27815680 DOI: 10.1007/s12264-016-0075-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 09/02/2016] [Indexed: 10/20/2022] Open
Affiliation(s)
- Shunling Guo
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA.
| | - Brad A Palanski
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Cornelius Kloeck
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Chaitan Khosla
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA.,Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
39
|
|
40
|
Development of Potent and Selective Tissue Transglutaminase Inhibitors: Their Effect on TG2 Function and Application in Pathological Conditions. ACTA ACUST UNITED AC 2015; 22:1347-61. [DOI: 10.1016/j.chembiol.2015.08.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 08/04/2015] [Accepted: 08/18/2015] [Indexed: 01/03/2023]
|
41
|
Vriezinga SL, Schweizer JJ, Koning F, Mearin ML. Coeliac disease and gluten-related disorders in childhood. Nat Rev Gastroenterol Hepatol 2015; 12:527-36. [PMID: 26100369 DOI: 10.1038/nrgastro.2015.98] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gluten-related disorders such as coeliac disease, wheat allergy and noncoeliac gluten sensitivity are increasingly being diagnosed in children. Coeliac disease occurs frequently, affecting 1-3% of the Western population. The condition manifests at a very young age, more so in girls, and is related to the HLA genotype. Coeliac disease might be considered a public health problem and, as primary prevention is not possible, the debate on mass screening should be reopened. Wheat proteins, including gluten, are responsible for one of the most common food allergies in children: wheat allergy. Unlike coeliac disease and wheat allergy, noncoeliac gluten sensitivity is an unclear and controversial entity. These three gluten-related disorders are treated with a gluten-free diet. In coeliac disease, the diet should be strictly followed, whereas wheat allergy only requires wheat elimination and in noncoeliac gluten sensitivity occasional trials of gluten reintroduction can be done. A good diagnostic work-up is important for gluten-related disorders in childhood to avoid unnecessary restrictive diets in children. In this Review, we provide an overview of the pathogenesis, diagnosis and management of the most common gluten-related disorders in children.
Collapse
Affiliation(s)
- Sabine L Vriezinga
- Department of Paediatrics, Leiden University Medical Centre, Albinusdreef 2/PO 9600, 2300 RC Leiden, Netherlands
| | - Joachim J Schweizer
- Department of Paediatrics, Leiden University Medical Centre, Albinusdreef 2/PO 9600, 2300 RC Leiden, Netherlands
| | - Frits Koning
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Albinusdreef 2/PO 9600, 2300 RC Leiden, Netherlands
| | - M Luisa Mearin
- Department of Paediatrics, Leiden University Medical Centre, Albinusdreef 2/PO 9600, 2300 RC Leiden, Netherlands
| |
Collapse
|
42
|
Costes LMM, Meresse B, Cerf-Bensussan N, Samsom JN. The role of animal models in unravelling therapeutic targets in coeliac disease. Best Pract Res Clin Gastroenterol 2015; 29:437-50. [PMID: 26060108 DOI: 10.1016/j.bpg.2015.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 04/21/2015] [Accepted: 04/26/2015] [Indexed: 01/31/2023]
Abstract
Coeliac disease is a complex small intestinal enteropathy that develops consequently to a breach of tolerance to gliadin, a storage protein abundantly found in cereals such as wheat, rye and barley. The understanding of the mechanisms underlying the development of coeliac disease in HLA-DQ2 and HLA-DQ8 genetically susceptible individuals has greatly improved during the last decades but so far did not allow to develop curative therapeutics, leaving a long-life gluten free diet as the only treatment option for the patients. In order to bring new therapeutic targets to light and to test the safety and efficacy of putative drugs, animal models recapitulating features of the disease are needed. Here, we will review the existing animal models and the clinical features of coeliac disease they reflect and discuss their relevance for modelling immune pathways that may lead to potential therapeutic approaches.
Collapse
Affiliation(s)
- Léa M M Costes
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus University Medical Center Rotterdam-Sophia Children's Hospital, Rotterdam, The Netherlands.
| | - Bertrand Meresse
- INSERM UMR1163, Laboratory of Intestinal Immunity, Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France.
| | - Nadine Cerf-Bensussan
- INSERM UMR1163, Laboratory of Intestinal Immunity, Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France.
| | - Janneke N Samsom
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus University Medical Center Rotterdam-Sophia Children's Hospital, Rotterdam, The Netherlands.
| |
Collapse
|
43
|
Plugis NM, Khosla C. Therapeutic approaches for celiac disease. Best Pract Res Clin Gastroenterol 2015; 29:503-21. [PMID: 26060114 PMCID: PMC4465084 DOI: 10.1016/j.bpg.2015.04.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/23/2015] [Accepted: 04/26/2015] [Indexed: 02/06/2023]
Abstract
Celiac disease is a common, lifelong autoimmune disorder for which dietary control is the only accepted form of therapy. A strict gluten-free diet is burdensome to patients and can be limited in efficacy, indicating there is an unmet need for novel therapeutic approaches to supplement or supplant dietary therapy. Many molecular events required for disease pathogenesis have been recently characterized and inspire most current and emerging drug-discovery efforts. Genome-wide association studies (GWAS) confirm the importance of human leukocyte antigen genes in our pathogenic model and identify a number of new risk loci in this complex disease. Here, we review the status of both emerging and potential therapeutic strategies in the context of disease pathophysiology. We conclude with a discussion of how genes identified during GWAS and follow-up studies that enhance susceptibility may offer insight into developing novel therapies.
Collapse
|