1
|
Kumar V, Xin X, Ma J, Tan C, Osna N, Mahato RI. Therapeutic targets, novel drugs, and delivery systems for diabetes associated NAFLD and liver fibrosis. Adv Drug Deliv Rev 2021; 176:113888. [PMID: 34314787 PMCID: PMC8440458 DOI: 10.1016/j.addr.2021.113888] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/12/2021] [Accepted: 07/18/2021] [Indexed: 02/08/2023]
Abstract
Type 2 diabetes mellitus (T2DM) associated non-alcoholic fatty liver disease (NAFLD) is the fourth-leading cause of death. Hyperglycemia induces various complications, including nephropathy, cirrhosis and eventually hepatocellular carcinoma (HCC). There are several etiological factors leading to liver disease development, which involve insulin resistance and oxidative stress. Free fatty acid (FFA) accumulation in the liver exerts oxidative and endoplasmic reticulum (ER) stresses. Hepatocyte injury induces release of inflammatory cytokines from Kupffer cells (KCs), which are responsible for activating hepatic stellate cells (HSCs). In this review, we will discuss various molecular targets for treating chronic liver diseases, including homeostasis of FFA, lipid metabolism, and decrease in hepatocyte apoptosis, role of growth factors, and regulation of epithelial-to-mesenchymal transition (EMT) and HSC activation. This review will also critically assess different strategies to enhance drug delivery to different cell types. Targeting nanocarriers to specific liver cell types have the potential to increase efficacy and suppress off-target effects.
Collapse
Affiliation(s)
- Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Xiaofei Xin
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jingyi Ma
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, University, MS 38677, USA
| | - Natalia Osna
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
2
|
Vascular adhesion protein-1 defines a unique subpopulation of human hematopoietic stem cells and regulates their proliferation. Cell Mol Life Sci 2021; 78:7851-7872. [PMID: 34719737 PMCID: PMC8629906 DOI: 10.1007/s00018-021-03977-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/30/2021] [Accepted: 10/12/2021] [Indexed: 10/25/2022]
Abstract
Although the development of hematopoietic stem cells (HSC) has been studied in great detail, their heterogeneity and relationships to different cell lineages remain incompletely understood. Moreover, the role of Vascular Adhesion Protein-1 in bone marrow hematopoiesis has remained unknown. Here we show that VAP-1, an adhesin and a primary amine oxidase producing hydrogen peroxide, is expressed on a subset of human HSC and bone marrow vasculature forming a hematogenic niche. Bulk and single-cell RNAseq analyses reveal that VAP-1+ HSC represent a transcriptionally unique small subset of differentiated and proliferating HSC, while VAP-1- HSC are the most primitive HSC. VAP-1 generated hydrogen peroxide acts via the p53 signaling pathway to regulate HSC proliferation. HSC expansion and differentiation into colony-forming units are enhanced by inhibition of VAP-1. Contribution of VAP-1 to HSC proliferation was confirmed with mice deficient of VAP-1, mice expressing mutated VAP-1 and using an enzyme inhibitor. In conclusion, VAP-1 expression allows the characterization and prospective isolation of a new subset of human HSC. Since VAP-1 serves as a check point-like inhibitor in HSC differentiation, the use of VAP-1 inhibitors enables the expansion of HSC.
Collapse
|
3
|
Forgács V, Németh E, Gyuricza B, Kis A, Szabó JP, Mikecz P, Mátyus P, Helyes Z, Horváth ÁI, Kálai T, Trencsényi G, Fekete A, Szikra D. Radiosynthesis and Preclinical Investigation of 11 C-Labelled 3-(4,5-Diphenyl-1,3-oxazol-2-yl)propanal Oxime ([ 11 C]SZV 1287). ChemMedChem 2020; 15:2470-2476. [PMID: 32935925 DOI: 10.1002/cmdc.202000389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/18/2020] [Indexed: 12/11/2022]
Abstract
The radiosynthesis, as well as the in vivo and ex vivo biodistribution of the 11 C radiolabelled 3-(4,5-diphenyl-1,3-oxazol-2-yl)propanal oxime (6, [11 C]SZV 1287) are reported. SZV 1287 is a novel semicarbazide-sensitive amine oxidase (SSAO) inhibitor and a promising candidate to be a novel analgesic for the treatment of neuropathic pain. Its radiolabelling was developed via a four-step radiosynthesis which started from the reaction of a Grignard reagent with [11 C]CO2 to produce [11 C]oxaprozin (3). In the next step this carboxylic acid 3 was directly reduced to yield the corresponding aldehyde, which was then converted into the oxime. [11 C]SZV 1287 was administered to male NMRI mice. The animals were examined with dynamic PET/MR imaging for 90 minutes. Biodistribution studies were performed at 10, 30, 60 and 120 minutes post injection. The accumulation of the labelled compound was observed in the brain of the animals. The main excretion pathway was found to be through the liver and intestines. These studies provide preliminary information for pharmacokinetic characterization of the SZV 1287.
Collapse
Affiliation(s)
- Viktória Forgács
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary.,Doctoral School of Chemistry, Faculty of Science and Technology, University of Debrecen, Egyetem tér 1., 4032, Debrecen, Hungary
| | - Enikő Németh
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary
| | - Barbara Gyuricza
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary.,Doctoral School of Chemistry, Faculty of Science and Technology, University of Debrecen, Egyetem tér 1., 4032, Debrecen, Hungary
| | - Adrienn Kis
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary.,Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary
| | - Judit P Szabó
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary.,Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary
| | - Pál Mikecz
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary
| | - Péter Mátyus
- Institute of Digital Health Sciences, Faculty of Health and Public Administration, Semmelweis University, Ferenc tér 15., 1094, Budapest, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12., 7624, Pécs, Hungary.,Molecular Pharmacology Research Group & Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, Szigeti str. 12., 7624, Pécs, Hungary
| | - Ádám István Horváth
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti str. 12., 7624, Pécs, Hungary.,Molecular Pharmacology Research Group & Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, Szigeti str. 12., 7624, Pécs, Hungary
| | - Tamás Kálai
- Institute of Organic and Medicinal Chemistry, Medical School, University of Pécs, Szigeti str. 12., 7624, Pécs, Hungary.,MTA-PTE Chronic Pain Research Group, Szigeti str. 12., 7624, Pécs, Hungary
| | - György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary
| | - Anikó Fekete
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary
| | - Dezső Szikra
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary
| |
Collapse
|
4
|
Romauch M. Zinc-α2-glycoprotein as an inhibitor of amine oxidase copper-containing 3. Open Biol 2020; 10:190035. [PMID: 32315567 PMCID: PMC6685929 DOI: 10.1098/rsob.190035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 07/04/2019] [Indexed: 12/12/2022] Open
Abstract
Zinc-α2-glycoprotein (ZAG) is a major plasma protein whose levels increase in chronic energy-demanding diseases and thus serves as an important clinical biomarker in the diagnosis and prognosis of the development of cachexia. Current knowledge suggests that ZAG mediates progressive weight loss through β-adrenergic signalling in adipocytes, resulting in the activation of lipolysis and fat mobilization. Here, through cross-linking experiments, amine oxidase copper-containing 3 (AOC3) is identified as a novel ZAG binding partner. AOC3-also known as vascular adhesion protein 1 (VAP-1) and semicarbazide sensitive amine oxidase (SSAO)-deaminates primary amines, thereby generating the corresponding aldehyde, H2O2 and NH3. It is an ectoenzyme largely expressed by adipocytes and induced in endothelial cells during inflammation. Extravasation of immune cells depends on amine oxidase activity and AOC3-derived H2O2 has an insulinogenic effect. The observations described here suggest that ZAG acts as an allosteric inhibitor of AOC3 and interferes with the associated pro-inflammatory and anti-lipolytic functions. Thus, inhibition of the deamination of lipolytic hormone octopamine by AOC3 represents a novel mechanism by which ZAG might stimulate lipolysis. Furthermore, experiments involving overexpression of recombinant ZAG reveal that its glycosylation is co-regulated by oxygen availability and that the pattern of glycosylation affects its inhibitory potential. The newly identified protein interaction between AOC3 and ZAG highlights a previously unknown functional relationship, which may be relevant to inflammation, energy metabolism and the development of cachexia.
Collapse
Affiliation(s)
- Matthias Romauch
- Institute of Molecular Biosciences, Karl-Franzens-University, Graz, Austria
| |
Collapse
|
5
|
Johnson BM, Shu YZ, Zhuo X, Meanwell NA. Metabolic and Pharmaceutical Aspects of Fluorinated Compounds. J Med Chem 2020; 63:6315-6386. [PMID: 32182061 DOI: 10.1021/acs.jmedchem.9b01877] [Citation(s) in RCA: 372] [Impact Index Per Article: 74.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The applications of fluorine in drug design continue to expand, facilitated by an improved understanding of its effects on physicochemical properties and the development of synthetic methodologies that are providing access to new fluorinated motifs. In turn, studies of fluorinated molecules are providing deeper insights into the effects of fluorine on metabolic pathways, distribution, and disposition. Despite the high strength of the C-F bond, the departure of fluoride from metabolic intermediates can be facile. This reactivity has been leveraged in the design of mechanism-based enzyme inhibitors and has influenced the metabolic fate of fluorinated compounds. In this Perspective, we summarize the literature associated with the metabolism of fluorinated molecules, focusing on examples where the presence of fluorine influences the metabolic profile. These studies have revealed potentially problematic outcomes with some fluorinated motifs and are enhancing our understanding of how fluorine should be deployed.
Collapse
Affiliation(s)
- Benjamin M Johnson
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Yue-Zhong Shu
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb Company, Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Xiaoliang Zhuo
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Nicholas A Meanwell
- Discovery Chemistry Platforms, Small Molecule Drug Discovery, Bristol Myers Squibb Company, Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| |
Collapse
|
6
|
Vakal S, Jalkanen S, Dahlström KM, Salminen TA. Human Copper-Containing Amine Oxidases in Drug Design and Development. Molecules 2020; 25:E1293. [PMID: 32178384 PMCID: PMC7144023 DOI: 10.3390/molecules25061293] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 12/28/2022] Open
Abstract
Two members of the copper-containing amine oxidase family are physiologically important proteins: (1) Diamine oxidase (hDAO; AOC1) with a preference for diamines is involved in degradation of histamine and (2) Vascular adhesion protein-1 (hVAP-1; AOC3) with a preference for monoamines is a multifunctional cell-surface receptor and an enzyme. hVAP-1-targeted inhibitors are designed to treat inflammatory diseases and cancer, whereas the off-target binding of the designed inhibitors to hDAO might result in adverse drug reactions. The X-ray structures for both human enzymes are solved and provide the basis for computer-aided inhibitor design, which has been reported by several research groups. Although the putative off-target effect of hDAO is less studied, computational methods could be easily utilized to avoid the binding of VAP-1-targeted inhibitors to hDAO. The choice of the model organism for preclinical testing of hVAP-1 inhibitors is not either trivial due to species-specific binding properties of designed inhibitors and different repertoire of copper-containing amine oxidase family members in mammalian species. Thus, the facts that should be considered in hVAP-1-targeted inhibitor design are discussed in light of the applied structural bioinformatics and structural biology approaches.
Collapse
Affiliation(s)
- Serhii Vakal
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Tykistökatu 6A, FI-20520 Turku, Finland; (S.V.); (K.M.D.)
| | - Sirpa Jalkanen
- MediCity Research Laboratory, University of Turku, Tykistökatu 6A, FI-20520 Turku, Finland;
| | - Käthe M. Dahlström
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Tykistökatu 6A, FI-20520 Turku, Finland; (S.V.); (K.M.D.)
| | - Tiina A. Salminen
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Tykistökatu 6A, FI-20520 Turku, Finland; (S.V.); (K.M.D.)
| |
Collapse
|
7
|
Papukashvili D, Rcheulishvili N, Deng Y. Attenuation of Weight Gain and Prevention of Associated Pathologies by Inhibiting SSAO. Nutrients 2020; 12:E184. [PMID: 31936548 PMCID: PMC7019322 DOI: 10.3390/nu12010184] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 12/19/2022] Open
Abstract
Obesity is a worldwide prevalent metabolic disorder that is associated with diabetes, among many other diseases. Bearing this in mind, prevention and treatment ways need to be improved. Notably, activity of the enzyme semicarbazide-sensitive amine oxidase (SSAO) is found to be elevated in overweight subjects. Moreover, SSAO inhibition has resulted in an increase of histamine activity in adipose tissue and the limitation of body fat. The current review aims to overview the risks of obesity, rationalize the molecular ways of SSAO activity, and outline the strategies of inhibiting upregulated enzyme levels. It describes the differences between SSAO inhibitors and advances the prospective agents. Based on evidence, caffeine is proposed as an effective, safe, and reliable choice to inhibit SSAO activity. Furthermore, the histamine in adipocytes has been associated with SSAO activity. Therefore, it is suggested as one of the key compounds to be studied for obesity management. To conclude, inhibiting SSAO may attenuate weight gain and prevent related diseases.
Collapse
Affiliation(s)
- Dimitri Papukashvili
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (D.P.); (N.R.)
| | - Nino Rcheulishvili
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (D.P.); (N.R.)
| | - Yulin Deng
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (D.P.); (N.R.)
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing 100081, China
| |
Collapse
|
8
|
Mercader J, Sabater AG, Le Gonidec S, Decaunes P, Chaplin A, Gómez-Zorita S, Milagro FI, Carpéné C. Oral Phenelzine Treatment Mitigates Metabolic Disturbances in Mice Fed a High-Fat Diet. J Pharmacol Exp Ther 2019; 371:555-566. [PMID: 31270215 DOI: 10.1124/jpet.119.259895] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022] Open
Abstract
Novel mechanisms and health benefits have been recently suggested for the antidepressant drug phenelzine (PHE), known as a nonselective monoamine oxidase inhibitor. They include an antilipogenic action that could have an impact on excessive fat accumulation and obesity-related metabolic alterations. We evaluated the metabolic effects of an oral PHE treatment on mice fed a high-fat diet (HFD). Eleven-week-old male C57BL/6 mice were fed a HFD and either a 0.028% PHE solution (HFD + PHE) or water to drink for 11 weeks. PHE attenuated the increase in body weight and adiposity without affecting food consumption. Energy efficiency was lower in HFD + PHE mice. Lipid content was reduced in subcutaneous fat pads, liver, and skeletal muscle. In white adipose tissue (WAT), PHE reduced sterol regulatory element-binding protein-1c and phosphoenolpyruvate carboxykinase mRNA levels, inhibited amine-induced lipogenesis, and did not increase lipolysis. Moreover, HFD + PHE mice presented diminished levels of hydrogen peroxide release in subcutaneous WAT and reduced expression of leukocyte transmigration markers and proinflammatory cytokines in visceral WAT and liver. PHE reduced the circulating levels of glycerol, triacylglycerols, high-density lipoprotein cholesterol, and insulin. Insulin resistance was reduced, without affecting glucose levels and glucose tolerance. In contrast, PHE increased rectal temperature and slightly increased energy expenditure. The mitigation of HFD-induced metabolic disturbances points toward a promising role for PHE in obesity treatment and encourages further research on its mechanisms of action. SIGNIFICANCE STATEMENT: Phenelzine reduces body fat, markers of oxidative stress, inflammation, and insulin resistance in high-fat diet mice. Semicarbazide-sensitive amine oxidase, monoamine oxidase, phosphoenolpyruvate carboxykinase, and sterol regulatory element-binding protein-1c are involved in the metabolic effects of phenelzine. Phenelzine could be potentially used for the treatment of obesity-related complications.
Collapse
Affiliation(s)
- Josep Mercader
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Agustín G Sabater
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Sophie Le Gonidec
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Pauline Decaunes
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Alice Chaplin
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Saioa Gómez-Zorita
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Fermín I Milagro
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| | - Christian Carpéné
- Balearic Islands Health Research Institute, Palma de Mallorca, Spain (J.M.); Department of Fundamental Biology and Health Sciences, University of Balearic Islands (UIB), Palma de Mallorca, Spain (J.M.); Alimentómica, S.L., Spin-off from UIB, Palma de Mallorca, Spain (A.G.S.); Institute of Metabolic and Cardiovascular Diseases, INSERM, UMR1048, Teams 1 & 3, Toulouse, France (C.C., S.L.G., P.D.); I2MC, University of Toulouse, UMR1048, Paul Sabatier University, Toulouse Cedex 4, France (C.C., S.L.G., P.D.); Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio (A.C.); Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country and Lucio Lascaray Research Institute, Vitoria, Spain (S.G.-Z.); CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain (S.G.-Z., F.I.M.); Department of Nutrition, Food Science, and Physiology, University of Navarra, Pamplona, Spain (F.I.M.); and Centre for Nutrition Research, University of Navarra, Pamplona, Spain (F.I.M.)
| |
Collapse
|
9
|
Findlay AD, Foot JS, Buson A, Deodhar M, Jarnicki AG, Hansbro PM, Liu G, Schilter H, Turner CI, Zhou W, Jarolimek W. Identification and Optimization of Mechanism-Based Fluoroallylamine Inhibitors of Lysyl Oxidase-like 2/3. J Med Chem 2019; 62:9874-9889. [DOI: 10.1021/acs.jmedchem.9b01283] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Alison D. Findlay
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, New South Wales 2086, Australia
| | - Jonathan S. Foot
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, New South Wales 2086, Australia
| | - Alberto Buson
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, New South Wales 2086, Australia
| | - Mandar Deodhar
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, New South Wales 2086, Australia
| | - Andrew G. Jarnicki
- Centre for Healthy Lungs, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales 2300, Australia
| | - Philip M. Hansbro
- Centre for Healthy Lungs, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales 2300, Australia
- Centre for Inflammation, Centenary Institute, Sydney, New South Wales 2050, Australia
- Faculty of Science, University of Technology Sydney, Ultimo, New South Wales 2007, Australia
| | - Gang Liu
- Centre for Inflammation, Centenary Institute, Sydney, New South Wales 2050, Australia
- Faculty of Science, University of Technology Sydney, Ultimo, New South Wales 2007, Australia
| | - Heidi Schilter
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, New South Wales 2086, Australia
| | - Craig I. Turner
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, New South Wales 2086, Australia
| | - Wenbin Zhou
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, New South Wales 2086, Australia
| | - Wolfgang Jarolimek
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, New South Wales 2086, Australia
| |
Collapse
|
10
|
Abstract
Significance: Vascular adhesion protein-1 (VAP-1) is an ectoenzyme that oxidates primary amines in a reaction producing also hydrogen peroxide. VAP-1 on the blood vessel endothelium regulates leukocyte extravasation from the blood into tissues under physiological and pathological conditions. Recent Advances: Inhibition of VAP-1 by neutralizing antibodies and by several novel small-molecule enzyme inhibitors interferes with leukocyte trafficking and alleviates inflammation in many experimental models. Targeting of VAP-1 also shows beneficial effects in several other diseases, such as ischemia/reperfusion, fibrosis, and cancer. Moreover, soluble VAP-1 levels may serve as a new prognostic biomarker in selected diseases. Critical Issues: Understanding the contribution of the enzyme activity-independent and enzyme activity-dependent functions, which often appear to be mediated by the hydrogen peroxide production, in the VAP-1 biology will be crucial. Similarly, there is a pressing need to understand which of the VAP-1 functions are regulated through the modulation of leukocyte trafficking, and what is the role of VAP-1 synthesized in adipose and smooth muscle cells. Future Directions: The specificity and selectivity of new VAP-1 inhibitors, and their value in animal models under therapeutic settings need to be addressed. Results from several programs studying the therapeutic potential of VAP-1 inhibition, which now are in clinical trials, will reveal the relevance of this amine oxidase in humans.
Collapse
Affiliation(s)
- Marko Salmi
- 1 MediCity , Turku, Finland .,2 Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sirpa Jalkanen
- 1 MediCity , Turku, Finland .,2 Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
11
|
Shanahan P, O'Sullivan J, Tipton KF, Kinsella GK, Ryan BJ, Henehan GTM. Theobromine and related methylxanthines as inhibitors of Primary Amine Oxidase. J Food Biochem 2018; 43:e12697. [PMID: 31353656 DOI: 10.1111/jfbc.12697] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 08/31/2018] [Accepted: 09/13/2018] [Indexed: 02/02/2023]
Abstract
Methylxanthines are among the most widely consumed drugs in the world and evidence of their health benefits has been growing in recent years. Primary Amine Oxidase (PrAO) has been recognized as a therapeutic target for the amelioration of inflammatory, vascular, and neurodegenerative diseases. Previous work in our laboratories showed that caffeine inhibited Bovine PrAO with a Ki of 1.0 mM using benzylamine as substrate. This study aimed to extend our previous work and explore the possibility that related methylxanthines might influence PrAO activity. While paraxanthine, theophylline, and 7-methylxanthine had little effect on PrAO, theobromine was a noncompetitive inhibitor with a Ki of 276 ± 44 µM. The specific structural elements of methylxanthines that are required for inhibition allow us to suggest that their binding site on PrAO may be a target for therapeutics. The health benefits associated with dietary methylxanthine consumption could involve PrAO inhibition. PRACTICAL APPLICATIONS: Inhibition of PrAO by methylxanthines may be significant in conferring health benefits. The design of PrAO inhibitors based on the structural motifs identified in this study (N-methylation at specific locations) is indicated. Existing therapeutics based on a core xanthine structure can be evaluated for their effects on PrAO. PrAO inhibition must be considered as a potential mediator of the beneficial health effects of some methylxanthines. If inhibition in human tissues is comparable to, or greater than, that found in these studies it points to an important role for these compounds in human health.
Collapse
Affiliation(s)
- Padraig Shanahan
- Applied Enzymology Group, School of Food Science and Environmental Health, College of Science and Health, Dublin Institute of Technology, Dublin, Ireland
| | | | - Keith F Tipton
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Gemma K Kinsella
- Applied Enzymology Group, School of Food Science and Environmental Health, College of Science and Health, Dublin Institute of Technology, Dublin, Ireland
| | - Barry J Ryan
- Applied Enzymology Group, School of Food Science and Environmental Health, College of Science and Health, Dublin Institute of Technology, Dublin, Ireland
| | - Gary T M Henehan
- Applied Enzymology Group, School of Food Science and Environmental Health, College of Science and Health, Dublin Institute of Technology, Dublin, Ireland
| |
Collapse
|
12
|
Horváth Á, Tékus V, Bencze N, Szentes N, Scheich B, Bölcskei K, Szőke É, Mócsai A, Tóth-Sarudy É, Mátyus P, Pintér E, Helyes Z. Analgesic effects of the novel semicarbazide-sensitive amine oxidase inhibitor SZV 1287 in mouse pain models with neuropathic mechanisms: Involvement of transient receptor potential vanilloid 1 and ankyrin 1 receptors. Pharmacol Res 2018; 131:231-243. [DOI: 10.1016/j.phrs.2018.02.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 02/07/2018] [Accepted: 02/07/2018] [Indexed: 12/11/2022]
|
13
|
Elo P, Tadayon S, Liljenbäck H, Teuho J, Käkelä M, Koskensalo K, Saunavaara V, Virta J, Veres TZ, Kiviniemi A, Saraste A, Marjamäki P, Airas L, Jalkanen S, Roivainen A. Vascular adhesion protein-1 is actively involved in the development of inflammatory lesions in rat models of multiple sclerosis. J Neuroinflammation 2018; 15:128. [PMID: 29716612 PMCID: PMC5930736 DOI: 10.1186/s12974-018-1152-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 04/09/2018] [Indexed: 11/16/2022] Open
Abstract
Background Vascular adhesion protein-1 (VAP-1) is an inflammation-inducible endothelial cell molecule and primary amine oxidase that mediates leukocyte entry to sites of inflammation. However, there is limited knowledge of the inflammation-related expression of VAP-1 in the central nervous system (CNS). Therefore, we investigated the expression of VAP-1 within the CNS vasculature in two focal rat models of experimental autoimmune encephalomyelitis (EAE) mimicking multiple sclerosis (MS). Methods EAE was induced either with Bacillus Calmette-Guérin, resulting in a delayed-type hypersensitivity-like pathogenesis (fDTH-EAE), or with myelin oligodendrocyte glycoprotein (fMOG-EAE). A subgroup of fMOG-EAE rats were treated daily with a selective VAP-1 inhibitor (LJP1586; 5 mg/kg). On 3 and 14 days after lesion activation, rat brains were assessed using magnetic resonance imaging (MRI), and ex vivo autoradiography was conducted to evaluate the binding of Gallium-68-labelled VAP-1 ligand. Histology and immunohistochemistry (OX-42, VAP-1, intercellular adhesion protein-1 [ICAM-1], P-selectin) supported the ex vivo autoradiography. Results EAE lesions showed MRI-detectable signal changes and binding of the VAP-1-targeting radiotracer in both rat models. Some of the VAP-1 positive vessels showed morphological features typical for high endothelial-like venules at sites of inflammation. Inhibition of VAP-1 activity with small molecule inhibitor, LJP1586, decreased lymphocyte density in the acute inflammatory phase of fMOG-EAE lesions (day 3, P = 0.026 vs. untreated), but not in the remission phase (day 14, P = 0.70 vs. untreated), and had no effect on the amount of OX-42-positive cells in either phase. LJP1586 treatment reduced VAP-1 and ICAM-1 expression in the acute inflammatory phase, whereas P-selectin remained not detectable at all studied stages of the disease. Conclusions Our results revealed that VAP-1 is expressed and functionally active in vasculature within the induced focal EAE lesions during the acute phase of inflammation and remains expressed after the acute inflammation has subsided. The study indicates that VAP-1 is actively involved in the development of inflammatory CNS lesions. During this process, the endothelial cell lesion-related vasculature seem to undergo a structural transformation from regular flat-walled endothelium to HEV-like endothelium.
Collapse
Affiliation(s)
- Petri Elo
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Sina Tadayon
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland.,Department of Radiology, Medical Imaging Centre of Southwest Finland, Turku University Hospital, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland
| | - Jarmo Teuho
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland.,Turku PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Meeri Käkelä
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Kalle Koskensalo
- Turku PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Virva Saunavaara
- Turku PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland.,Department of Medical physics, Turku University Hospital, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Jenni Virta
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Tibor Z Veres
- MediCity Research Laboratory, University of Turku, Tykistökatu 6, FI-20520, Turku, Finland
| | - Aida Kiviniemi
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland.,Turku PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland.,Department of Radiology, Medical Imaging Centre of Southwest Finland, Turku University Hospital, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Antti Saraste
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Päivi Marjamäki
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Laura Airas
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland.,Division of Clinical Neurosciences, Turku University Hospital, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland
| | - Sirpa Jalkanen
- MediCity Research Laboratory, University of Turku, Tykistökatu 6, FI-20520, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland. .,Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland. .,Turku PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland.
| |
Collapse
|
14
|
Zhang Y, Yi W, Yao J, Yu X, Qian C, Hu Z. Hypoxia serves a key function in the upregulated expression of vascular adhesion protein‑1 in vitro and in a rat model of hemorrhagic shock. Mol Med Rep 2017. [PMID: 28627649 PMCID: PMC5562078 DOI: 10.3892/mmr.2017.6727] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hemorrhagic shock following major trauma results in mortality, but the function of vascular adhesion protein-1 (VAP-1), implicated in intracranial hemorrhage, remains unknown. This study aimed to determine whether expression of the AOC3 gene and its encoded protein, VAP-1, is altered by hypoxia. Rat hepatic sinusoidal endothelial cells (RHSECs) and rat intestinal microvascular endothelial cells (RIMECs) were transduced with a viral vector carrying AOC3, and AOC3 mRNA expression levels were measured by reverse transcription-quantitative polymerase chain reaction. VAP-1 protein expression levels were measured by western blot analysis and compared between normoxic and hypoxic conditions. Following this, AOC3 mRNA and VAP-1 protein expression levels in hepatic and intestinal tissues were assessed in a rat model of hemorrhagic shock with or without fluid resuscitation; and serum semicarbazide-sensitive amine oxidase (SSAO) activity was measured by fluorometric assays. The effects of 2-bromoethylamine (2-BEA) on AOC3/VAP-1 levels and 24 h survival were investigated. AOC3 mRNA and VAP-1 protein levels were increased in RHSECs and RIMECs by hypoxia, and in hepatic and intestinal tissues from rats following hemorrhagic shock. Hypoxia increased serum SSAO activity in these animals. 2-BEA reduced AOC3 mRNA and VAP-1 protein levels in hepatic and intestinal tissues from rats following hemorrhagic shock, and appeared to improve survival in animals not receiving resuscitation following hemorrhagic shock. In conclusion, hemorrhagic shock upregulates AOC3/VAP-1 expressions, and this potentially occurs via hypoxia. Therefore, inhibition of VAP-1 may be beneficial in the setting of hemorrhagic shock. Further studies are required to confirm these findings and to establish whether VAP-1 may be a valid target for the development of novel therapies for hemorrhagic shock.
Collapse
Affiliation(s)
- Yuxing Zhang
- Department of General Surgery, Navy General Hospital, Beijing 100048, P.R. China
| | - Wei Yi
- Department of General Surgery, China People's Liberation Army No. 94 Hospital, Nanchang, Jiangxi 330002, P.R. China
| | - Jun Yao
- Department of General Surgery, Shanghai Changzheng Hospital, Shanghai 200003, P.R. China
| | - Xiaojun Yu
- Department of Gastroenterological Surgery, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Cheng Qian
- Department of General Surgery, Huzhou Maternity & Child Care Hospital, Huzhou, Zhejiang 313000, P.R. China
| | - Zhiqian Hu
- Department of General Surgery, Shanghai Changzheng Hospital, Shanghai 200003, P.R. China
| |
Collapse
|
15
|
Tanaka S, Tanaka T, Kawakami T, Takano H, Sugahara M, Saito H, Higashijima Y, Yamaguchi J, Inagi R, Nangaku M. Vascular adhesion protein-1 enhances neutrophil infiltration by generation of hydrogen peroxide in renal ischemia/reperfusion injury. Kidney Int 2017; 92:154-164. [PMID: 28318627 DOI: 10.1016/j.kint.2017.01.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 12/28/2016] [Accepted: 01/05/2017] [Indexed: 10/19/2022]
Abstract
Vascular adhesion protein-1 (VAP-1) is a unique molecule since it acts as an adhesion molecule as well as an ectoenzyme catalyzing oxidative deamination of primary amines and generates hydrogen peroxide in the extracellular space. While VAP-1 is implicated in various inflammatory diseases, its role in acute kidney injury is less characterized. Here we studied VAP-1 expression in the kidney and the effect of its inhibition in a rat model of renal ischemia/reperfusion injury. VAP-1 was predominantly expressed in pericytes, which released enzymatically active enzyme. In vivo, a specific VAP-1 inhibitor, RTU-1096, significantly ameliorated rat renal ischemia/reperfusion injury and decreased neutrophil infiltration measured 12 hours after injury without altering macrophage or T lymphocyte populations. The protective effect of VAP-1 inhibition was lost in neutrophil-depleted rats, suggesting its inhibition ameliorated renal ischemia/reperfusion injury by suppressing neutrophil infiltration. To investigate whether hydrogen peroxide generated by VAP-1 enzyme reaction enhances neutrophil infiltration, we conducted an under-agarose migration assay with purified human neutrophils. Recombinant human VAP-1 significantly induced neutrophil migration, which was almost completely inhibited by RTU-1096 or catalase. Thus, VAP-1 plays a critical role in the pathophysiology of renal ischemia/reperfusion injury by enhancement of neutrophil infiltration generating a local hydrogen peroxide gradient. Hence, VAP-1 inhibition may be a novel therapy in ischemic acute kidney injury.
Collapse
Affiliation(s)
- Shinji Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takahisa Kawakami
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Hideki Takano
- Division of Nephrology, Tokyo Teishin Hospital, Tokyo, Japan
| | - Mai Sugahara
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Hisako Saito
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yoshiki Higashijima
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Junna Yamaguchi
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Reiko Inagi
- Division of Chronic Kidney Disease Pathophysiology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
16
|
Marttila-Ichihara F, Elima K, Auvinen K, Veres TZ, Rantakari P, Weston C, Miyasaka M, Adams D, Jalkanen S, Salmi M. Amine oxidase activity regulates the development of pulmonary fibrosis. FASEB J 2017; 31:2477-2491. [PMID: 28251930 DOI: 10.1096/fj.201600935r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 02/07/2017] [Indexed: 12/19/2022]
Abstract
In pulmonary fibrosis, an inflammatory reaction and differentiation of myofibroblasts culminate in pathologic deposition of collagen. Amine oxidase copper containing-3 (AOC3) is a cell-surface-expressed oxidase that regulates leukocyte extravasation. Here we analyzed the potential role of AOC3 using gene-modified and inhibitor-treated mice in a bleomycin-induced pulmonary fibrosis model. Inflammation and fibrosis of lungs were assessed by histologic, flow cytometric, and quantitative PCR analysis. AOC3-deficient mice showed a 30-50% reduction in fibrosis, collagen synthesis, numbers of myofibroblasts, and accumulation of CD4+ lymphocytes, NK T cells, macrophages, and type 2 innate lymphoid cells compared with wild-type control mice. AOC3-knock-in mice, which express a catalytically inactive form of AOC3, were also protected from lung fibrosis. In wild-type mice, a small-molecule AOC3 inhibitor treatment reduced leukocyte infiltration, myofibroblast differentiation, and fibrotic injury both in prophylactic and early therapeutic settings by about 50% but was unable to reverse the established fibrosis. AOC3 was also induced in myofibroblasts in human idiopathic pulmonary fibrosis. Thus, the oxidase activity of AOC3 contributes to the development of lung fibrosis mainly by regulating the accumulation of pathogenic leukocyte subtypes, which drive the fibrotic response.-Marttila-Ichihara, F., Elima, K., Auvinen, K., Veres, T. Z., Rantakari, P., Weston, C., Miyasaka, M., Adams, D., Jalkanen, S., Salmi, M. Amine oxidase activity regulates the development of pulmonary fibrosis.
Collapse
Affiliation(s)
| | - Kati Elima
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
| | - Kaisa Auvinen
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Tibor Z Veres
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Pia Rantakari
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Christopher Weston
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Unit, University of Birmingham, Birmingham, United Kingdom; and
| | - Masayuki Miyasaka
- MediCity Research Laboratory, University of Turku, Turku, Finland.,World Premier International (WPI) Immunology Frontier Research Center, Osaka University, Japan
| | - David Adams
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Unit, University of Birmingham, Birmingham, United Kingdom; and
| | - Sirpa Jalkanen
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Marko Salmi
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| |
Collapse
|
17
|
Analgesic and Anti-Inflammatory Effects of the Novel Semicarbazide-Sensitive Amine-Oxidase Inhibitor SzV-1287 in Chronic Arthritis Models of the Mouse. Sci Rep 2017; 7:39863. [PMID: 28067251 PMCID: PMC5220351 DOI: 10.1038/srep39863] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 11/28/2016] [Indexed: 01/03/2023] Open
Abstract
Semicarbazide-sensitive amine oxidase (SSAO) catalyses oxidative deamination of primary amines. Since there is no data about its function in pain and arthritis mechanisms, we investigated the effects of our novel SSAO inhibitor SzV-1287 in chronic mouse models of joint inflammation. Effects of SzV-1287 (20 mg/kg i.p./day) were investigated in the K/BxN serum-transfer and complete Freund's adjuvant (CFA)-evoked active immunization models compared to the reference SSAO inhibitor LJP-1207. Mechanonociception was assessed by aesthesiometry, oedema by plethysmometry, clinical severity by scoring, joint function by grid test, myeloperoxidase activity by luminescence, vascular leakage by fluorescence in vivo imaging, histopathological changes by semiquantitative evaluation, and cytokines by Luminex assay. SzV-1287 significantly inhibited hyperalgesia and oedema in both models. Plasma leakage and keratinocyte chemoattractant production in the tibiotarsal joint, but not myeloperoxidase activity was significantly reduced by SzV-1287 in K/BxN-arthritis. SzV-1287 did not influence vascular and cellular mechanisms in CFA-arthritis, but significantly decreased histopathological alterations. There was no difference in the anti-hyperalgesic and anti-inflammatory actions of SzV-1287 and LJP-1207, but only SzV-1287 decreased CFA-induced tissue damage. Unlike SzV-1287, LJP-1207 induced cartilage destruction, which was confirmed in vitro. SzV-1287 exerts potent analgesic and anti-inflammatory actions in chronic arthritis models of distinct mechanisms, without inducing cartilage damage.
Collapse
|
18
|
Leukocyte trafficking-associated vascular adhesion protein 1 is expressed and functionally active in atherosclerotic plaques. Sci Rep 2016; 6:35089. [PMID: 27731409 PMCID: PMC5059718 DOI: 10.1038/srep35089] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/23/2016] [Indexed: 12/15/2022] Open
Abstract
Given the important role of inflammation and the potential association of the leukocyte trafficking-associated adhesion molecule vascular adhesion protein 1 (VAP-1) with atherosclerosis, this study examined whether functional VAP-1 is expressed in atherosclerotic lesions and, if so, whether it could be targeted by positron emission tomography (PET). First, immunohistochemistry revealed that VAP-1 localized to endothelial cells of intra-plaque neovessels in human carotid endarterectomy samples from patients with recent ischemic symptoms. In low-density lipoprotein receptor-deficient mice expressing only apolipoprotein B100 (LDLR-/-ApoB100/100), VAP-1 was expressed on endothelial cells lining inflamed atherosclerotic lesions; normal vessel walls in aortas of C57BL/6N control mice were VAP-1-negative. Second, we discovered that the focal uptake of VAP-1 targeting sialic acid-binding immunoglobulin-like lectin 9 based PET tracer [68Ga]DOTA-Siglec-9 in atherosclerotic plaques was associated with the density of activated macrophages (r = 0.58, P = 0.022). As a final point, we found that the inhibition of VAP-1 activity with small molecule LJP1586 decreased the density of macrophages in inflamed atherosclerotic plaques in mice. Our results suggest for the first time VAP-1 as a potential imaging target for inflamed atherosclerotic plaques, and corroborate VAP-1 inhibition as a therapeutic approach in the treatment of atherosclerosis.
Collapse
|
19
|
Botz B, Bölcskei K, Helyes Z. Challenges to develop novel anti-inflammatory and analgesic drugs. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [PMID: 27576790 DOI: 10.1002/wnan.1427] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/21/2016] [Accepted: 07/30/2016] [Indexed: 12/11/2022]
Abstract
Chronic inflammatory diseases and persistent pain of different origin represent common medical, social, and economic burden, and their pharmacotherapy is still an unresolved issue. Therefore, there is a great and urgent need to develop anti-inflammatory and analgesic agents with novel mechanisms of action, but it is a very challenging task. The main problem is the relatively large translational gap between the preclinical experimental data and the clinical results due to characteristics of the models, difficulties with the investigational techniques particularly for pain, as well as species differences in the mechanisms. We summarize here the current state-of-the-art medication and related ongoing strategies, and the novel targets with lead molecules under clinical development. The first members of the gold-standard categories, such as nonsteroidal anti-inflammatory drugs, glucocorticoids, and opioids, were introduced decades ago, and since then very few drugs with novel mechanisms of action have been successfully taken to the clinics despite considerable development efforts. Several biologics targeting different key molecules have provided breakthrough in some autoimmune/inflammatory diseases, but they are expensive, only parenterally available, their long-term side effects often limit their administration, and they do not effectively reduce pain. Some kinase inhibitors and phosphodiesterase-4 blockers have recently been introduced as new directions. There are in fact some promising novel approaches at different clinical stages of drug development focusing on transient receptor potential vanilloid 1/ankyrin 1 channel antagonism, inhibition of voltage-gated sodium/calcium channels, several enzymes (kinases, semicarbazide-sensitive amine oxidases, and matrix metalloproteinases), cytokines/chemokines, transcription factors, nerve growth factor, and modulation of several G protein-coupled receptors (cannabinoids, purinoceptors, and neuropeptides). WIREs Nanomed Nanobiotechnol 2017, 9:e1427. doi: 10.1002/wnan.1427 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Bálint Botz
- Department of Radiology, Faculty of Medicine, University of Pécs, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Kata Bölcskei
- János Szentágothai Research Centre, University of Pécs, Pécs, Hungary.,Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Pécs, Pécs, Hungary
| | - Zsuzsanna Helyes
- János Szentágothai Research Centre, University of Pécs, Pécs, Hungary.,Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Pécs, Pécs, Hungary.,MTA-PTE NAP B Chronic Pain Research Group, Faculty of Medicine, University of Pécs, Pécs, Hungary
| |
Collapse
|
20
|
Expression of the semicarbazide-sensitive amine oxidase in articular cartilage: its role in terminal differentiation of chondrocytes in rat and human. Osteoarthritis Cartilage 2016; 24:1223-34. [PMID: 26851450 DOI: 10.1016/j.joca.2016.01.340] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 01/15/2016] [Accepted: 01/20/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Semicarbazide-sensitive amine oxidase (SSAO) catalyzes the oxidation of primary amines into ammonia and reactive species (hydrogen peroxide, aldehydes). It is highly expressed in mammalian tissues, especially in vascular smooth muscle cells and adipocytes, where it plays a role in cell differentiation and glucose transport. The study aims at characterizing the expression and the activity of SSAO in rat and human articular cartilage of the knee, and to investigate its potential role in chondrocyte terminal differentiation. DESIGN SSAO expression was examined by immunohistochemistry and western blot. Enzyme activity was measured using radiolabeled benzylamine as a substrate. Primary cell cultures of rat chondrocytes were treated for 21 days by a specific SSAO inhibitor, LJP 1586. Terminal chondrocyte differentiation markers were quantified by RT-qPCR. The basal and IL1β-stimulated glucose transport was monitored by the entrance of (3)[H]2-deoxyglucose in chondrocytes. RESULTS SSAO was expressed in chondrocytes of rat and human articular cartilage. SSAO expression was significantly enhanced during the hypertrophic differentiation of chondrocytes characterized by an increase in MMP13 and in alkaline phosphatase (ALP) expressions. SSAO inhibition delayed the late stage of chondrocyte differentiation without cell survival alteration and diminished the basal and IL1β-stimulated glucose transport. Interestingly, SSAO activity was strongly increased in human osteoarthritic cartilage. CONCLUSIONS SSAO was expressed as an active form in rat and human cartilage. The results suggest the involvement of SSAO in rat chondrocyte terminal differentiation via a modulation of the glucose transport. In man, the increased SSAO activity detected in osteoarthritic patients may trigger hypertrophy and cartilage degeneration.
Collapse
|
21
|
Interstitial renal fibrosis due to multiple cisplatin treatments is ameliorated by semicarbazide-sensitive amine oxidase inhibition. Kidney Int 2016; 89:374-85. [DOI: 10.1038/ki.2015.327] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 08/31/2015] [Accepted: 09/03/2015] [Indexed: 02/07/2023]
|
22
|
Pannecoeck R, Serruys D, Benmeridja L, Delanghe JR, van Geel N, Speeckaert R, Speeckaert MM. Vascular adhesion protein-1: Role in human pathology and application as a biomarker. Crit Rev Clin Lab Sci 2015; 52:284-300. [PMID: 26287391 DOI: 10.3109/10408363.2015.1050714] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Vascular adhesion protein-1 (VAP-1) is a member of the copper-containing amine oxidase/semicarbazide-sensitive amine oxidase (AOC/SSAO) enzyme family. SSAO enzymes catalyze oxidative deamination of primary amines, which results in the production of the corresponding aldehyde, hydrogen peroxide and ammonium. VAP-1 is continuously expressed as a transmembrane glycoprotein in the vascular wall during development and facilitates the accumulation of inflammatory cells into the inflamed environment in concert with other leukocyte adhesion molecules. The soluble form of VAP-1 is released into the circulation mainly from vascular endothelial cells. Over- and under-expression of sVAP-1 result in alterations of the reported reaction product levels, which are involved in the pathogenesis of multiple human diseases. The combination of enzymatic and adhesion capacities as well as its strong association with inflammatory pathologies makes VAP-1 an interesting therapeutic target for drug discovery. In this article, we will review the general characteristics and biological functions of VAP-1, focusing on its important role as a prognostic biomarker in human pathologies. In addition, the potential therapeutic application of VAP-1 inhibitors will be discussed.
Collapse
Affiliation(s)
| | | | | | | | - Nanja van Geel
- c Department of Dermatology , Ghent University Hospital , Gent , Belgium
| | | | | |
Collapse
|
23
|
Xu H, Testai FD, Valyi-Nagy T, N Pavuluri M, Zhai F, Nanegrungsunk D, Paisansathan C, Pelligrino DA. VAP-1 blockade prevents subarachnoid hemorrhage-associated cerebrovascular dilating dysfunction via repression of a neutrophil recruitment-related mechanism. Brain Res 2015; 1603:141-9. [PMID: 25662771 DOI: 10.1016/j.brainres.2015.01.047] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 01/19/2015] [Accepted: 01/27/2015] [Indexed: 11/19/2022]
Abstract
Our previous findings indicated that in rats subjected to subarachnoid hemorrhage (SAH), suppression of post-SAH neuroinflammation via vascular adhesion protein-1 (VAP-1) blockade provides significant neuroprotection. We and others have reported that neuroinflammation contributes to cerebral microvascular impairment. Thus, in the present study, we tested the hypotheses that: (1) treatment with LJP-1586, a selective VAP-1 blocker, prevents SAH-associated pial arteriolar dilating dysfunction; and (2) the vasculoprotective effect of LJP-1586 arises from inhibiting SAH-elicited neutrophil recruitment. We utilized an endovascular perforation model of SAH. Rats subjected to SAH were either treated with LJP-1586 or rendered neutropenic via anti-neutrophil-antibody treatment. Findings from these groups were compared to their respective control groups. At 48 h post-SAH, rats were evaluated for neurobehavioral function, pial venular leukocyte trafficking, and pial arteriolar reactivity to topically-applied acetylcholine (ACh) and S-nitroso-N-acetyl penicillamine (SNAP). Pial arteriolar responses decreased at 48 h post-SAH. However, in the presence of LJP-1586, those responses were significantly preserved. Neutrophil-depletion yielded a substantial suppression of SAH-associated leukocyte adhesion and infiltration. This was accompanied by a significant preservation of pial arteriolar dilating function, suggesting a direct link between neutrophil recruitment and the loss of cerebral microvascular reactivity. Moreover, neutrophil depletion also was associated with significant protection of neurobehavioral function. The present findings suggest that attenuating SAH-linked elevation in neutrophil trafficking will protect against the development of microvascular dysfunction and subsequent neurological impairment.
Collapse
Affiliation(s)
- Haoliang Xu
- Neuroanesthesia Research Laboratory, Department of Anesthesiology, University of Illinois-Chicago, Chicago, IL, United States.
| | - Fernando D Testai
- Department of Neurology and Rehabilitation, University of Illinois-Chicago, Chicago, IL, United States
| | - Tibor Valyi-Nagy
- Department of Pathology, University of Illinois-Chicago, Chicago, IL, United States
| | - Mani N Pavuluri
- Department of Psychiatry, University of Illinois-Chicago, Chicago, IL, United States
| | - Fengguo Zhai
- Neuroanesthesia Research Laboratory, Department of Anesthesiology, University of Illinois-Chicago, Chicago, IL, United States
| | - Danop Nanegrungsunk
- Neuroanesthesia Research Laboratory, Department of Anesthesiology, University of Illinois-Chicago, Chicago, IL, United States
| | - Chanannait Paisansathan
- Neuroanesthesia Research Laboratory, Department of Anesthesiology, University of Illinois-Chicago, Chicago, IL, United States
| | - Dale A Pelligrino
- Neuroanesthesia Research Laboratory, Department of Anesthesiology, University of Illinois-Chicago, Chicago, IL, United States
| |
Collapse
|
24
|
Wong M, Saad S, Zhang J, Gross S, Jarolimek W, Schilter H, Chen JA, Gill AJ, Pollock CA, Wong MG. Semicarbazide-sensitive amine oxidase (SSAO) inhibition ameliorates kidney fibrosis in a unilateral ureteral obstruction murine model. Am J Physiol Renal Physiol 2014; 307:F908-16. [DOI: 10.1152/ajprenal.00698.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Semicarbazide-sensitive amine oxidase (SSAO) is an enzyme known for its dual function in mediating inflammation and reactive oxygen species production. However, the role of SSAO inhibitors in limiting kidney fibrosis is unclear. We aimed to determine the effectiveness of a SSAO inhibitor (SSAOi; PXS4728A) as an antifibrotic agent using a 7-day unilateral ureteric obstruction (UUO) model of acute kidney fibrosis in 6- to 8-wk-old mice. The experimental groups were 1) Sham operated; 2) UUO; 3) UUO+SSAOi (2 mg/kg); 4) UUO+telmisartan, an angiotensin receptor blocker (3 mg/kg); and 5) UUO+SSAOi+telmisartan. Kidney tissue was analyzed for histological evidence of tubulointerstitial fibrosis, nitrotyrosine staining, and mRNA expression of markers associated with fibrosis and inflammation. Kidney SSAO activity was determined by radiometric [14C]benzylamine methodology. Our results show that SSAOi effectively suppresses UUO-mediated SSAO activity. Extracellular matrix markers, namely, fibronectin, collagen IV protein, and nitrotyrosine staining, were lower in UUO+SSAOi mice compared with untreated UUO mice. This was consistent with the attenuated mRNA expression of collagen IV and fibronectin. SSAOi effectively inhibited transforming growth factor-β1 (TGF-β1) and monocyte chemoattractant protein-1 (MCP-1) expression to a similar extent to that observed with telmisartan. Individually, SSAOi and telmisartan induced a reduction in interstitial leukocyte and macrophage accumulation. However, the combination of SSAOi and telmisartan was more effective at reducing inflammatory cell infiltration. These results demonstrate that SSAO inhibition significantly suppresses profibrotic and proinflammatory cytokine secretion, reduces oxidative stress, and limits inflammatory cell accumulation and extracellular matrix expression in an acute model of renal fibrosis.
Collapse
Affiliation(s)
- May Wong
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, St. Leonards, Sydney, New South Wales, Australia
| | - Sonia Saad
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, St. Leonards, Sydney, New South Wales, Australia
| | - Jie Zhang
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, St. Leonards, Sydney, New South Wales, Australia
| | - Simon Gross
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, St. Leonards, Sydney, New South Wales, Australia
| | - Wolfgang Jarolimek
- Pharmaxis, Limited, Frenchs Forest, Sydney, New South Wales, Australia; and
| | - Heidi Schilter
- Pharmaxis, Limited, Frenchs Forest, Sydney, New South Wales, Australia; and
| | - Jason A. Chen
- Department of Anatomical Pathology, Royal North Shore Hospital, University of Sydney, St. Leonards, Sydney, New South Wales, Australia
| | - Anthony J. Gill
- Department of Anatomical Pathology, Royal North Shore Hospital, University of Sydney, St. Leonards, Sydney, New South Wales, Australia
| | - Carol A. Pollock
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, St. Leonards, Sydney, New South Wales, Australia
| | - Muh Geot Wong
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, St. Leonards, Sydney, New South Wales, Australia
| |
Collapse
|
25
|
Huang TC, Chen SM, Li YC, Lee JA. Increased renal semicarbazide-sensitive amine oxidase activity and methylglyoxal levels in aristolochic acid-induced nephrotoxicity. Life Sci 2014; 114:4-11. [DOI: 10.1016/j.lfs.2014.07.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 07/03/2014] [Accepted: 07/25/2014] [Indexed: 02/08/2023]
|
26
|
Finney J, Moon HJ, Ronnebaum T, Lantz M, Mure M. Human copper-dependent amine oxidases. Arch Biochem Biophys 2014; 546:19-32. [PMID: 24407025 DOI: 10.1016/j.abb.2013.12.022] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/24/2013] [Accepted: 12/26/2013] [Indexed: 12/11/2022]
Abstract
Copper amine oxidases (CAOs) are a class of enzymes that contain Cu(2+) and a tyrosine-derived quinone cofactor, catalyze the conversion of a primary amine functional group to an aldehyde, and generate hydrogen peroxide and ammonia as byproducts. These enzymes can be classified into two non-homologous families: 2,4,5-trihydroxyphenylalanine quinone (TPQ)-dependent CAOs and the lysine tyrosylquinone (LTQ)-dependent lysyl oxidase (LOX) family of proteins. In this review, we will focus on recent developments in the field of research concerning human CAOs and the LOX family of proteins. The aberrant expression of these enzymes is linked to inflammation, fibrosis, tumor metastasis/invasion and other diseases. Consequently, there is a critical need to understand the functions of these proteins at the molecular level, so that strategies targeting these enzymes can be developed to combat human diseases.
Collapse
Affiliation(s)
- Joel Finney
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
| | - Hee-Jung Moon
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
| | - Trey Ronnebaum
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
| | - Mason Lantz
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
| | - Minae Mure
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
27
|
Foot JS, Yow TT, Schilter H, Buson A, Deodhar M, Findlay AD, Guo L, McDonald IA, Turner CI, Zhou W, Jarolimek W. PXS-4681A, a potent and selective mechanism-based inhibitor of SSAO/VAP-1 with anti-inflammatory effects in vivo. J Pharmacol Exp Ther 2013; 347:365-74. [PMID: 23943052 DOI: 10.1124/jpet.113.207613] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Semicarbazide-sensitive amine oxidase (SSAO), also known as vascular adhesion protein-1 (VAP-1), is a member of the copper-dependent amine oxidase family that is associated with various forms of inflammation and fibrosis. To investigate the therapeutic potential of SSAO/VAP-1 inhibition, potent and selective inhibitors with drug-like properties are required. PXS-4681A [(Z)-4-(2-(aminomethyl)-3-fluoroallyloxy)benzenesulfonamide hydrochloride] is a mechanism-based inhibitor of enzyme function with a pharmacokinetic and pharmacodynamic profile that ensures complete, long-lasting inhibition of the enzyme after a single low dose in vivo. PXS-4681A irreversibly inhibits the enzyme with an apparent Ki of 37 nM and a kinact of 0.26 min(-1) with no observed turnover in vitro. It is highly selective for SSAO/VAP-1 when profiled against related amine oxidases, ion channels, and seven-transmembrane domain receptors, and is superior to previously reported inhibitors. In mouse models of lung inflammation and localized inflammation, dosing of this molecule at 2 mg/kg attenuates neutrophil migration, tumor necrosis factor-α, and interleukin-6 levels. These results demonstrate the drug-like properties of PXS-4681A and its potential use in the treatment of inflammation.
Collapse
|
28
|
Wong MYW, Saad S, Pollock C, Wong MG. Semicarbazide-sensitive amine oxidase and kidney disease. Am J Physiol Renal Physiol 2013; 305:F1637-44. [PMID: 24173357 DOI: 10.1152/ajprenal.00416.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
With better understanding of the molecular mechanisms underpinning chronic kidney disease, the roles of inflammation and fibrosis are becoming increasingly inseparable. The progression of renal disease is characterized by pathomorphological changes that consist of early inflammatory responses followed by tubulointerstitial fibrosis, tubular atrophy, and glomerular and vascular sclerosis. Currently available therapies that reduce hypertension, proteinuria, hyperglycemia, and interruption of the renin-angiotensin-aldosterone system are at best only partially effective. Hence, there remains a need to explore agents targeting nonrenin-angiotensin-aldosterone system pathways. In this review, we discuss mechanistic aspects in the physiological and pathological role of semicarbazide-sensitive amine oxidase, a protein enzyme involved in cellular trafficking and inflammation, with respect to the kidney. We explore the evidence for the use of semicarbazide-sensitive amine oxidase inhibitors as potential agents in renal fibrosis to delay the onset and progression of chronic kidney disease.
Collapse
Affiliation(s)
- May Y W Wong
- Kolling Institute of Medical Research, Level 7 Kolling Bldg., Royal North Shore Hospital, St Leonards 2065, NSW, Australia.
| | | | | | | |
Collapse
|
29
|
Watcharotayangul J, Mao L, Xu H, Vetri F, Baughman VL, Paisansathan C, Pelligrino DA. Post-ischemic vascular adhesion protein-1 inhibition provides neuroprotection in a rat temporary middle cerebral artery occlusion model. J Neurochem 2012; 123 Suppl 2:116-24. [PMID: 23050649 DOI: 10.1111/j.1471-4159.2012.07950.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We examined the neuroprotective efficacy associated with post-ischemic vascular adhesion protein-1 (VAP-1) blockade in rats subjected to transient (1 h) middle cerebral artery occlusion (MCAo). We compared saline-treated control rats to rats treated with a highly selective VAP-1 inhibitor, LJP-1586 [Z-3-fluoro-2-(4-methoxybenzyl) allylamine hydrochloride]. Initial intraperitoneal LJP-1586 (or saline control) treatments were delayed until 6 h or 12 h reperfusion. At 72-h reperfusion, LJP-1586-treated rats displayed 51% and 33% smaller infarct volumes, relative to their controls, in the 6- and 12-h treatment groups, respectively. However, only in the 6-h treatment group was the infarct volume reduction significant (p < 0.05). On the other hand, we observed significantly improved neurologic functions in both 6- and 12-h treatment groups, versus their matched controls (p < 0.05). Also, the effect of 6-h LJP-1586 treatment on post-ischemic leukocyte trafficking in pial venules overlying the ischemic cortex was evaluated using intravital microscopy. These experiments revealed that: 1) LJP-1586 did not affect intravascular leukocyte (largely neutrophil) adhesion, at least out to 12-h reperfusion; and 2) the onset of neutrophil extravasation, which occurred between 6-8-h reperfusion in control rats, was prevented by LJP-1586-treatment. In conclusion, in rats subjected to transient MCAo, selective VAP-1 pharmacologic blockade provided neuroprotection, with a prolonged therapeutic window of 6-12-h reperfusion.
Collapse
|
30
|
Bergeron M, Guyader D, Paquin JF. S(N)2' reaction of allylic difluorides with lithium amides and thiolates. Org Lett 2012; 14:5888-91. [PMID: 23145465 DOI: 10.1021/ol302802r] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The synthesis of monofluoroalkenes using an S(N)2' reaction of lithium amides derived from aromatic amines or lithium thiolates with 3,3-difluoropropenes is reported. This transformation features the use of fluoride as a leaving group.
Collapse
Affiliation(s)
- Maxime Bergeron
- Canada Research Chair in Organic and Medicinal Chemistry, Département de chimie, Université Laval, 1045 avenue de la Médecine, Québec, Québec G1V 0A6, Canada
| | | | | |
Collapse
|
31
|
The discovery and development of selective 3-fluoro-4-aryloxyallylamine inhibitors of the amine oxidase activity of semicarbazide-sensitive amine oxidase/vascular adhesion protein-1 (SSAO/VAP-1). Bioorg Med Chem Lett 2012; 22:3935-40. [PMID: 22595173 DOI: 10.1016/j.bmcl.2012.04.111] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/18/2012] [Accepted: 04/23/2012] [Indexed: 11/21/2022]
Abstract
A new class of 3-fluoroallyl amine-based SSAO/VAP-1 inhibitors is reported. These compounds have excellent selectivity over diamine oxidase, MAO-A and MAO-B. Synthesis and SAR studies leading to compound 28 (PXS-4159A) are reported. The pharmacokinetic profile of 28 in the rat, together with activity in a murine model of lung inflammation are also disclosed.
Collapse
|
32
|
Carpéné C, Desquesnes A, Gomez-Ruiz A, Iffiú-Soltész Z, Le Gonidec S, Mercader J. Long-term activation of semicarbazide-sensitive amine oxidase lowers circulating levels of uric acid in diabetic conditions. Physiol Res 2012; 61:251-7. [PMID: 22480418 DOI: 10.33549/physiolres.932211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Uric acid is involved in nitrogenous waste in animals, together with ammonia and urea. Uric acid has also antioxidant properties and is a surrogate marker of metabolic syndrome. We observed that the elevated plasma uric acid of high-fat fed mice was normalized by benzylamine treatment. Indeed, benzylamine is the reference substrate of semicarbazide-sensitive amine oxidase (SSAO), an enzyme highly expressed in fat depots and vessels, which generates ammonia when catalysing oxidative deamination. Ammonia interferes with uric acid metabolism/solubility. Our aim was therefore to investigate whether the lowering action of benzylamine on uric acid was related to an improvement of diabetic complications, or was connected with SSAO-dependent ammonia production. First, we observed that benzylamine administration lowered plasma uric acid in diabetic db/db mice while it did not modify uric acid levels in normoglycemic and lean mice. In parallel, benzylamine improved the glycemic control in diabetic but not in normoglycemic mice, while plasma urea remained unaltered. Then, uric acid plasma levels were measured in mice invalidated for AOC3 gene, encoding for SSAO. These mice were unable to oxidize benzylamine but were not diabetic and exhibited unaltered plasma uric levels. Therefore, activated or abolished ammonia production by SSAO was without influence on uric acid in the context of normoglycemia. Our observations confirm that plasma uric acid increases with diabetes and can be normalized when glucose tolerance is improved. They also show that uric acid, a multifunctional metabolite at the crossroads of nitrogen waste and of antioxidant defences, can be influenced by SSAO, in a manner apparently related to changes in glucose homeostasis.
Collapse
Affiliation(s)
- C Carpéné
- INSERM U1048 équipe 3, I2MC, Bat. L4, CHU Rangueil, BP 84225, Toulouse Cedex 4, France.
| | | | | | | | | | | |
Collapse
|
33
|
Herder V, Hansmann F, Stangel M, Schaudien D, Rohn K, Baumgärtner W, Beineke A. Cuprizone inhibits demyelinating leukomyelitis by reducing immune responses without virus exacerbation in an infectious model of multiple sclerosis. J Neuroimmunol 2012; 244:84-93. [PMID: 22329906 DOI: 10.1016/j.jneuroim.2012.01.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Revised: 01/20/2012] [Accepted: 01/23/2012] [Indexed: 10/28/2022]
Abstract
Multiple sclerosis is one of the most common demyelinating central nervous system diseases in young adults. Theiler's murine encephalomyelitis (TME) is a widely used virus-induced murine model for human myelin disorders. Immunosuppressive approaches generally reduce antiviral immunity and therefore increase virus dissemination with clinical worsening. In the present study, the progressive course of TME was significantly delayed due to a five-week cuprizone feeding period. Cuprizone was able to minimize demyelinating leukomyelitis without virus exacerbation. This phenomenon is supposed to be a consequence of selective inhibition of detrimental inflammatory responses with maintained protective immunity against the virus.
Collapse
Affiliation(s)
- Vanessa Herder
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, D-30559 Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Peet GW, Lukas S, Hill-Drzewi M, Martin L, Rybina IV, Roma T, Shoultz A, Zhu X, Cazacu D, Kronkaitis A, Baptiste A, Raudenbush BC, August EM, Modis LK. Bioluminescent Method for Assaying Multiple Semicarbazide-Sensitive Amine Oxidase (SSAO) Family Members in Both 96- and 384-Well Formats. ACTA ACUST UNITED AC 2011; 16:1106-11. [DOI: 10.1177/1087057111414897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Vascular adhesion protein–1 (VAP-1), also known as semicarbazide-sensitive amine oxidase (SSAO) or copper-containing amine oxidase (AOC3, EC 1.4.3.6), catalyzes oxidative deamination of primary amines. One endogenous substrate has recently been described (Siglec 10), and although its mechanism of action in vivo is not completely understood, it is suggested to play a role in immune cell trafficking, making it a target of interest for autoimmune and inflammatory diseases. Much of the enzymology performed around this target has been conducted with absorbance, fluorescent, or radiometric formats that can have some limitations for high-throughput screening and subsequent compound profiling. The authors present the use of a bioluminescent assay, originally developed for monoamine oxidase enzymes, in a high-throughput format. It can be used for related SSAOs such as AOC1 given their substrate similarity with VAP-1. The authors also demonstrate that it is compatible with different sources of VAP-1, both purified recombinant and VAP-1 overexpressed on live cells.
Collapse
Affiliation(s)
| | - Susan Lukas
- Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | | | - Leslie Martin
- Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | | | - Teresa Roma
- Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Alycia Shoultz
- Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Xiang Zhu
- Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Daniela Cazacu
- Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | | | | | | | | | | |
Collapse
|
36
|
Siglec-9 is a novel leukocyte ligand for vascular adhesion protein-1 and can be used in PET imaging of inflammation and cancer. Blood 2011; 118:3725-33. [PMID: 21821708 DOI: 10.1182/blood-2010-09-311076] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leukocyte migration to sites of inflammation is regulated by several endothelial adhesion molecules. Vascular adhesion protein-1 (VAP-1) is unique among the homing-associated molecules as it is both an enzyme that oxidizes primary amines and an adhesin. Although granulocytes can bind to endothelium via a VAP-1-dependent manner, the counter-receptor(s) on this leukocyte population is(are) not known. Here we used a phage display approach and identified Siglec-9 as a candidate ligand on granulocytes. The binding between Siglec-9 and VAP-1 was confirmed by in vitro and ex vivo adhesion assays. The interaction sites between VAP-1 and Siglec-9 were identified by molecular modeling and confirmed by further binding assays with mutated proteins. Although the binding takes place in the enzymatic groove of VAP-1, it is only partially dependent on the enzymatic activity of VAP-1. In positron emission tomography, the ⁶⁸Gallium-labeled peptide of Siglec-9 specifically detected VAP-1 in vasculature at sites of inflammation and cancer. Thus, the peptide binding to the enzymatic groove of VAP-1 can be used for imaging conditions, such as inflammation and cancer.
Collapse
|
37
|
Lin Z, Li H, Luo H, Zhang Y, Luo W. Benzylamine and methylamine, substrates of semicarbazide-sensitive amine oxidase, attenuate inflammatory response induced by lipopolysaccharide. Int Immunopharmacol 2011; 11:1079-89. [PMID: 21414430 DOI: 10.1016/j.intimp.2011.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 02/17/2011] [Accepted: 03/01/2011] [Indexed: 02/05/2023]
Abstract
Current evidence indicates that semicarbazide-sensitive amine oxidase (SSAO) substrates possess insulin-mimic effect, which was thought to play an anti-inflammatory role. The purpose of the present study was to determine whether SSAO substrates benzylamine (BZA) and methylamine (MA) attenuate inflammatory response induced by lipopolysaccharide (LPS). BALB/c mice peritoneal macrophages (PMs) that express SSAO and RAW264.7 mouse macrophages that do not express SSAO were used in vitro studies. Experimental mice were given BZA or MA through intraperitoneal injection before LPS challenge. The results showed that BZA or MA treatment significantly reduced LPS-induced pro-inflammatory mediators (nitric oxide, TNF-α) production, the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2, and glucose consumption in murine PMs, but not in RAW264.7 cell line. The metabolites of BZA or MA catalyzed by SSAO, hydrogen peroxide, formaldehyde, and benzaldehyde could also significantly decrease LPS-induced nitric oxide and TNF-α production, iNOS and COX-2 expression, and glucose consumption in vitro. In addition, BZA or MA administration could significantly decrease plasma pro-inflammatory mediators and the expression of iNOS and COX-2 in liver and lung, and could also attenuate LPS-induced transient hyperglycemia and chronic hypoglycemia. These findings indicated that substrates of SSAO might be involved in the anti-inflammatory effects. The metabolites of BZA and MA catalyzed by SSAO might be responsible for the anti-inflammatory effects. Moreover, BZA or MA administration could be useful for normalization of glucose disposal during endotoxemia.
Collapse
Affiliation(s)
- Zhexuan Lin
- Bio-analytical Laboratory, Shantou University Medical College, Shantou, PR China
| | | | | | | | | |
Collapse
|
38
|
Weston CJ, Adams DH. Hepatic consequences of vascular adhesion protein-1 expression. J Neural Transm (Vienna) 2011; 118:1055-64. [PMID: 21512782 DOI: 10.1007/s00702-011-0647-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Accepted: 04/10/2011] [Indexed: 01/09/2023]
Abstract
The liver is constantly exposed to antigens present in the blood and to particulate antigens delivered from the gut. To maintain effective levels of immune surveillance and yet tolerate food antigens, the hepatic environment has become highly specialised. A low flow environment exists within the hepatic sinusoids that not only facilitates the exchange of toxins and nutrients within the liver parenchyma, but also provides an ideal niche for the recruitment of leukocytes. One such adhesion molecule involved in this process, the vascular adhesion protein-1 (VAP-1), is unusual in the context of the leukocyte adhesion cascade in that it is both an adhesion molecule and a primary amine oxidase. In this review, we examine the biological functions of VAP-1 and examine what role this molecule might play in the establishment and progression of chronic liver disease.
Collapse
Affiliation(s)
- Chris J Weston
- Centre for Liver Research and NIHR Biomedical Research Unit, 5th Floor, Institute of Biomedical Research, MRC Centre for Immune Regulation, College of Medicine and Dentistry, University of Birmingham, Birmingham, Edgbaston, B15 2TT, UK.
| | | |
Collapse
|
39
|
Dunkel P, Balogh B, Meleddu R, Maccioni E, Gyires K, Mátyus P. Semicarbazide-sensitive amine oxidase/vascular adhesion protein-1: a patent survey. Expert Opin Ther Pat 2011; 21:1453-71. [PMID: 21675926 DOI: 10.1517/13543776.2011.594040] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Vascular adhesion protein-1 (VAP-1)/semicarbazide-sensitive amine oxidase (SSAO) is an adhesion protein involved in leukocyte trafficking and inflammatory processes, with a special amine oxidase activity. Inhibitors have been mainly developed for treating chronic inflammatory disorders. The utility of inhibitors as antiangiogenic agents in ophthalmological and oncological diseases is currently under evaluation. SSAO substrates may mimic several insulin effects, although their utility for the treatment of diabetes is still far from being fully understood. AREAS COVERED This paper reviews the patent literature of SSAO/VAP-1 inhibitors and substrates, for the period of 1990 - 2010. The current stage of SSAO/VAP-1-interacting agents published in patents is described, along with their chemical structures and pharmacological uses. EXPERT OPINION SSAO/VAP-1 is a promising anti-inflammatory target. Another important field for therapeutic application of these inhibitors may be ophthalmology, due to their antiangiogenic effects. SSAO substrates might also be of therapeutic value in the treatment of diabetes; however, more extensive research has to be undertaken to validate this approach.
Collapse
Affiliation(s)
- Petra Dunkel
- Semmelweis University, Department of Organic Chemistry , Hőgyes Endre utca 7, 1092 Budapest , Hungary
| | | | | | | | | | | |
Collapse
|
40
|
Salmi M, Jalkanen S. Homing-associated molecules CD73 and VAP-1 as targets to prevent harmful inflammations and cancer spread. FEBS Lett 2011; 585:1543-50. [DOI: 10.1016/j.febslet.2011.04.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 04/13/2011] [Accepted: 04/14/2011] [Indexed: 01/01/2023]
|
41
|
Vascular adhesion protein-1 inhibition provides antiinflammatory protection after an intracerebral hemorrhagic stroke in mice. J Cereb Blood Flow Metab 2011; 31:881-93. [PMID: 20877383 PMCID: PMC3063621 DOI: 10.1038/jcbfm.2010.167] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The systemic immune response has a vital role in propagating the damage of an intracerebral hemorrhage (ICH). Vascular adhesion protein-1 (VAP-1), a semicarbazide (SCZ)-sensitive-amine-oxidase, was found in previous studies to have a role in migration of immune cells. In this study, we hypothesize that VAP-1 inhibition may decrease brain injury by attenuating the transmigration of immune cells to the injury site, and by doing so, reduce cerebral edema and improve neurobehavioral function in mice. Two VAP-1 inhibitors, LJP1586 and SCZ were given 1 hour after ICH induction by either collagenase or autologous blood injection. The VAP-1 siRNA, a VAP-1 gene silencer, and human recombinant AOC3 protein, a VAP-1 analogue, were delivered by intracerebroventricular injection. Postassessment included neurobehavioral testing, brain edema measurement, quantification of neutrophil infiltration and microglia/macrophage activation, and measurement of intercellular adhesion molecule-1 (ICAM-1), P-selectin, monocyte chemoattractant protein-1 (MCP-1), and tumor necrosis factor-α (TNF-α) expression 24 hours after ICH. We found that LJP1586 and SCZ reduced brain edema and neurobehavioral deficits 24 hours after ICH induction. These two drugs were also found to decrease levels of ICAM-1, MCP-1, TNF-α, and inhibit neutrophilic infiltration and microglia/macrophage activation. We conclude that VAP-1 inhibition provided antiinflammation effect by reducing adhesion molecule expression and immune cell infiltration after ICH.
Collapse
|
42
|
Marttila-Ichihara F, Castermans K, Auvinen K, Oude Egbrink MGA, Jalkanen S, Griffioen AW, Salmi M. Small-molecule inhibitors of vascular adhesion protein-1 reduce the accumulation of myeloid cells into tumors and attenuate tumor growth in mice. THE JOURNAL OF IMMUNOLOGY 2010; 184:3164-73. [PMID: 20154208 DOI: 10.4049/jimmunol.0901794] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Vascular adhesion protein-1 (VAP-1) is an endothelial, cell surface-expressed oxidase involved in leukocyte traffic. The adhesive function of VAP-1 can be blocked by anti-VAP-1 Abs and small-molecule inhibitors. However, the effects of VAP-1 blockade on antitumor immunity and tumor progression are unknown. In this paper, we used anti-VAP-1 mAbs and small-molecule inhibitors of VAP-1 in B16 melanoma and EL-4 lymphoma tumor models in C57BL/6 mice. Leukocyte accumulation into tumors and neoangiogenesis were evaluated by immunohistochemistry, flow cytometry, and intravital videomicroscopy. We found that both anti-VAP-1 Abs and VAP-1 inhibitors reduced the number of leukocytes in the tumors, but they targeted partially different leukocyte subpopulations. Anti-VAP-1 Abs selectively inhibited infiltration of CD8-positive lymphocytes into tumors and had no effect on accumulation of myeloid cells into tumors. In contrast, the VAP-1 inhibitors significantly reduced only the number of proangiogenic Gr-1(+)CD11b(+) myeloid cells in melanomas and lymphomas. Blocking of VAP-1 by either means left tumor homing of regulatory T cells and type 2 immune-suppressing monocytes/macrophages intact. Notably, VAP-1 inhibitors, but not anti-VAP-1 Abs, retarded the growth of melanomas and lymphomas and reduced tumor neoangiogenesis. The VAP-1 inhibitors also reduced the binding of Gr-1(+) myeloid cells to the tumor vasculature. We conclude that tumors use the catalytic activity of VAP-1 to recruit myeloid cells into tumors and to support tumor progression. Small-molecule VAP-1 inhibitors therefore might be a potential new tool for immunotherapy of tumors.
Collapse
|
43
|
Largeron M, Fleury MB, Strolin Benedetti M. A small molecule that mimics the metabolic activity of copper-containing amine oxidases (CuAOs) toward physiological mono- and polyamines. Org Biomol Chem 2010; 8:3796-800. [DOI: 10.1039/c004501b] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
44
|
Kiss J, Jalkanen S, Fülöp F, Savunen T, Salmi M. Ischemia-reperfusion injury is attenuated in VAP-1-deficient mice and by VAP-1 inhibitors. Eur J Immunol 2009; 38:3041-9. [PMID: 18991279 DOI: 10.1002/eji.200838651] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Neutrophils mediate the damage caused by ischemia-reperfusion both at the site of primary injury and in remote organs. Vascular adhesion protein-1 (VAP-1) is an ectoenzyme expressed on endothelial cells and it has been shown to regulate leukocyte extravasation. Here we show for the first time using VAP-1-deficient mice that VAP-1 plays a significant role in the intestinal damage and acute lung injury after ischemia-reperfusion. Separate inhibition of VAP-1 by small molecule enzyme inhibitors and a function-blocking monoclonal antibody in WT mice revealed that the catalytic activity of VAP-1 is responsible for its pro-inflammatory action. The use of transgenic humanized VAP-1 mice also showed that the enzyme inhibitors alleviate both the ischemia-reperfusion injury in the gut and neutrophil accumulation in the lungs. These data thus indicate that VAP-1 regulates the inflammatory response in ischemia-reperfusion injury and suggest that blockade of VAP-1 may have therapeutic value.
Collapse
Affiliation(s)
- Jan Kiss
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | | | | | | | | |
Collapse
|
45
|
Martelius T, Salmi M, Krogerus L, Loginov R, Schoultz M, Karikoski M, Miiluniemi M, Soots A, Höckerstedt K, Jalkanen S, Lautenschlager I. Inhibition of Semicarbazide-Sensitive Amine Oxidases Decreases Lymphocyte Infiltration in the Early Phases of Rat Liver Allograft Rejection. Int J Immunopathol Pharmacol 2008; 21:911-20. [DOI: 10.1177/039463200802100415] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Vascular adhesion protein-1 (VAP-1) has been shown to mediate lymphocyte adhesion to endothelia at sites of inflammation in vitro and in vivo. VAP-1 is also an ectoenzyme with semicarbazide-sensitive amine oxidase (SSAO) activity. In this study we investigated whether inhibition of SSAO influences the inflammatory infiltration in acute rat liver allograft rejection. BN recipients of DA liver allografts were treated with 50 mg/kg/d semicarbazide, an inhibitor of SSAO, or similar volumes of saline. 10 rats/group were followed for graft survival, and 10 rats/group were sacrificed on day 7 post-transplantation for histology and T-lymphocyte isolation. The area percentage of portal inflammatory infiltrates in the grafts was assessed from digital photomicrographs. The proportion of CD4-, CD8- and IL2-receptor positive lymphocytes in the graft was quantified with flow cytometry. On day 7, semicarbazide treatment significantly decreased the inflammatory infiltrate area in the grafts. CD4-, CD8- and IL2-receptor positive cells were equally affected. However, animal survival was not affected. Blockade of the enzymatic activity of VAP-1 has a significant effect on lymphocyte infiltration early in acute liver rejection. Later, activation of other adhesion pathways can by-pass the blockade caused by VAP-inhibition.
Collapse
Affiliation(s)
- T. Martelius
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki
- Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki
| | - M. Salmi
- MediCity Research Laboratory, University of Turku, Turku
- Department of Bacterial and Inflammatory Diseases, National Public Health Institute, Turku, Finland
| | - L. Krogerus
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki
| | - R. Loginov
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki
- Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki
| | - M. Schoultz
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki
| | - M. Karikoski
- MediCity Research Laboratory, University of Turku, Turku
| | - M. Miiluniemi
- MediCity Research Laboratory, University of Turku, Turku
| | - A. Soots
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki
| | - K. Höckerstedt
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki
| | - S. Jalkanen
- MediCity Research Laboratory, University of Turku, Turku
- Department of Bacterial and Inflammatory Diseases, National Public Health Institute, Turku, Finland
| | - I. Lautenschlager
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki
- Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki
| |
Collapse
|