1
|
Wissbroecker KB, Zmuda AJ, Karumanchi H, Niehaus TD. Biochemical and genomic evidence for converging metabolic routes of metformin and biguanide breakdown in environmental Pseudomonads. J Biol Chem 2024; 300:107935. [PMID: 39476966 DOI: 10.1016/j.jbc.2024.107935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 12/01/2024] Open
Abstract
Metformin is commonly used to lower blood glucose levels and is one of the most widely used pharmaceuticals worldwide. Typical doses are high (0.5-2.0 g day-1) and the majority travels through the digestive system unabsorbed and enters the wastewater system. Metformin is not removed by standard wastewater treatments and eventually enters freshwater systems, where it can form N-chloro-derivatives that are toxic to fish and human cells. Thus, metformin is one of the most prevalent anthropogenic pollutants worldwide and there has been considerable interest in finding ways to remove it. We recently isolated Pseudomonads capable of growing on metformin as the sole nitrogen source. We identified candidate genes involved in metformin breakdown through genomic analyses informed by feeding studies. One candidate, a pair of genes that are located on ∼80kb extra-genomic plasmids, was shown to encode a heteromeric Ni-dependent hydrolase that converts metformin to guanylurea and dimethylamine. Metforminase activity of these gene products is now well established as our results confirm three recently published independent studies. Our isolated Pseudomonads also grow on biguanide, suggesting the existence of an additional breakdown enzyme. Another candidate gene located on the ∼80kb plasmids was shown to encode an aminohydrolase that converts biguanide to guanylurea. Biguanide may arise through successive N-demethylations of metformin or come from other sources. Our results suggest that the recent evolution of metforminase and biguanide hydrolase enzymes allow Pseudomonads to convert either metformin or biguanide to guanylurea, which can be assimilated by existing pathways.
Collapse
Affiliation(s)
- Katie B Wissbroecker
- The Department of Plant and Microbial Biology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anthony J Zmuda
- The Department of Plant and Microbial Biology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Harsheeth Karumanchi
- The Department of Plant and Microbial Biology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Thomas D Niehaus
- The Department of Plant and Microbial Biology, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
2
|
Contini L, Paul A, Mazzei L, Ciurli S, Roncarati D, Braga D, Grepioni F. Is bismuth(III) able to inhibit the activity of urease? Puzzling results in the quest for soluble urease complexes for agrochemical and medicinal applications. Dalton Trans 2024; 53:10553-10562. [PMID: 38847020 DOI: 10.1039/d4dt00778f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Bismuth(III) complexes have been reported to act as inhibitors of the enzyme urease, ubiquitously present in soils and implicated in the pathogenesis of several microorganisms. The general insolubility of Bi(III) complexes in water at neutral pH, however, is an obstacle to their utilization. In our quest to improve the solubility of Bi(III) complexes, we selected a compound reported to inhibit urease, namely [Bi(HEDTA)]·2H2O, and co-crystallized it with (i) racemic DL-histidine to obtain the conglomerate [Bi2(HEDTA)2(μ-D-His)2]·6H2O + [Bi2(HEDTA)2(μ-L-His)2]·6H2O, (ii) enantiopure L-histidine to yield [Bi2(HEDTA)2(μ-L-His)2]·6H2O, and (iii) cytosine to obtain [Bi(HEDTA)]·Cyt·2H2O. All compounds, synthesised by mechanochemical methods and by slurry, were characterized in the solid state by calorimetric (DSC and TGA) and spectroscopic (IR) methods, and their structures were determined using powder X-ray diffraction (PXRD) data. All compounds show an appreciable solubility in water, with values ranging from 6.8 mg mL-1 for the starting compound [Bi(HEDTA)]·2H2O to 36 mg mL-1 for [Bi2(HEDTA)2(μ-L-His)2]·6H2O. The three synthesized compounds as well as [Bi(HEDTA)]·2H2O were then tested for inhibition activity against urease. Surprisingly, no enzymatic inhibition was observed during in vitro assays using Canavalia ensiformis urease and in vivo assays using cultures of Helicobacter pylori, raising questions on the efficacy of Bi(III) compounds to counteract the negative effects of urease activity in the agro-environment and in human health.
Collapse
Affiliation(s)
- Laura Contini
- Department of Chemistry "G. Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy.
| | - Arundhati Paul
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Viale Giuseppe Fanin 40, Bologna I-40127, Italy.
| | - Luca Mazzei
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Viale Giuseppe Fanin 40, Bologna I-40127, Italy.
| | - Stefano Ciurli
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Viale Giuseppe Fanin 40, Bologna I-40127, Italy.
| | - Davide Roncarati
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Selmi 3, 40126 Bologna, Italy.
| | - Dario Braga
- Department of Chemistry "G. Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy.
| | - Fabrizia Grepioni
- Department of Chemistry "G. Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy.
| |
Collapse
|
3
|
Muller J, Marchisio L, Attia R, Zedet A, Maradan R, Vallet M, Aebischer A, Harakat D, Senejoux F, Ramseyer C, Foley S, Cardey B, Girard C, Pudlo M. A colorimetric assay adapted to fragment screening revealing aurones and chalcones as new arginase inhibitors. RSC Med Chem 2024; 15:1722-1730. [PMID: 38784454 PMCID: PMC11110760 DOI: 10.1039/d3md00713h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/23/2024] [Indexed: 05/25/2024] Open
Abstract
Arginase, a difficult-to-target metalloenzyme, is implicated in a wide range of diseases, including cancer, infectious, and cardiovascular diseases. Despite the medical need, existing inhibitors have limited structural diversity, consisting predominantly of amino acids and their derivatives. The search for innovative arginase inhibitors has now extended to screening approaches. Due to the small and narrow active site of arginase, screening must meet the criteria of fragment-based screening. However, the limited binding capacity of fragments requires working at high concentrations, which increases the risk of interference and false positives. In this study, we investigated three colorimetric assays and selected one based on interference for screening under these challenging conditions. The subsequent adaptation and application to the screening a library of metal chelator fragments resulted in the identification of four compounds with moderate activity. The synthesis and evaluation of a series of compounds from one of the hits led to compound 21a with an IC50 value of 91.1 μM close to the reference compound piceatannol. Finally, molecular modelling supports the potential binding of aurones and chalcones to the active site of arginase, suggesting them as new candidates for the development of novel arginase inhibitors.
Collapse
Affiliation(s)
- Jason Muller
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Luca Marchisio
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Rym Attia
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Andy Zedet
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Robin Maradan
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Maxence Vallet
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Alison Aebischer
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Dominique Harakat
- URCATech, ICMR, CNRS UMR 7312 URCA Bât 18, BP 1039, Cedex 2 51687 Reims France
| | - François Senejoux
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Christophe Ramseyer
- Chrono-environnement UMR6249, CNRS Université de Franche-Comté F-25000 Besançon France
| | - Sarah Foley
- Chrono-environnement UMR6249, CNRS Université de Franche-Comté F-25000 Besançon France
| | - Bruno Cardey
- Chrono-environnement UMR6249, CNRS Université de Franche-Comté F-25000 Besançon France
| | - Corine Girard
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| | - Marc Pudlo
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT F-25000 Besançon France
| |
Collapse
|
4
|
Ma H, Liu X, Wen Z, Yi X, Liu Y, Zhou H. Competitive Mn(II) removal occurs in Lysinibacillus sp. MHQ-1 through microbially-induced carbonate precipitation (MICP) and indirect Mn(II) oxidation. ENVIRONMENTAL RESEARCH 2023; 239:117373. [PMID: 37827367 DOI: 10.1016/j.envres.2023.117373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
Biological Mn(II) removal usually involves adsorption and precipitation of Mn(II) in the form of various minerals. Manganese oxides (MnOx) formation through the activity of Mn(II) oxidation bacteria (MnOB) contributes to the majority of Mn(II) removal. However, whether other bacterial-mediated pathway could couple or competitive with Mn(II) oxidation during Mn(II) removal is scarcely reported. In this study, we reported a competitive Mn(II) removal occurred in nutrient-rich condition during the indirect Mn(II) oxidation of Lysinibacillus sp. MHQ-1, i.e., microbially-induced carbonate precipitation (MICP). In the presence of 1 mM Mn(II), 39.4% of free Mn(II) converted to MnCO3(s) quickly within 100 h, and then 11.6% of initial Mn(II) slowly oxidized to MnOx within 442 h. The urease activity assay and the genome sequencing confirmed the existence of urease and the absence of Mn(II)-oxidizing enzymes in the genome of strain MHQ-1. The urease catalyzed the formation of carbonate ion that reacts with Mn(II) and the formed ammonia raises the pH to initiate indirect Mn(II) oxidation. Genome survey suggests the urease widely exists in various Mn(II)-oxidizing bacteria (MnOB), emphasizing the importance to reconsider the composition, stability and environmental effects of biological Mn(II) removal products in nutrient-rich environment.
Collapse
Affiliation(s)
- Huiqing Ma
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Ocean Science and Technology, Panjin Campus, Dalian University of Technology, China
| | - Xinyue Liu
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Ocean Science and Technology, Panjin Campus, Dalian University of Technology, China
| | - Zhaoqi Wen
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Ocean Science and Technology, Panjin Campus, Dalian University of Technology, China
| | - Xianliang Yi
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Ocean Science and Technology, Panjin Campus, Dalian University of Technology, China
| | - Yang Liu
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Ocean Science and Technology, Panjin Campus, Dalian University of Technology, China
| | - Hao Zhou
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), School of Ocean Science and Technology, Panjin Campus, Dalian University of Technology, China.
| |
Collapse
|
5
|
Xie Y, Zhang Y, Liu X, Cao L, Han M, Wang C, Chen J, Zhang X. miR‑151a‑5p promotes the proliferation and metastasis of colorectal carcinoma cells by targeting AGMAT. Oncol Rep 2023; 49:50. [PMID: 36704851 PMCID: PMC9887461 DOI: 10.3892/or.2023.8487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 01/11/2023] [Indexed: 01/22/2023] Open
Abstract
Colorectal carcinoma (CRC) is one of the most common types of digestive cancer. It has been reported that the ectopic expression of microRNAs (miRs) plays a critical role in the occurrence and progression of CRC. In addition, it has also been suggested that miR‑151a‑5p may serve as a useful biomarker for the early detection and treatment of different types of cancer and particularly CRC. However, the specific effects and underlying mechanisms of miR‑151a‑5p in CRC remain elusive. The results of the current study demonstrated that miR‑151a‑5p was upregulated in CRC cell lines and clinical tissues derived from patients with CRC. Functionally, the results showed that miR‑151a‑5p significantly promoted CRC cell proliferation, migration and invasion. Additionally, dual luciferase reporter assays verified that agmatinase (AGMAT) was a direct target of miR‑151a‑5p and it was positively associated with miR‑151a‑5p expression. Mechanistically, miR‑151a‑5p could enhance the epithelial‑mesenchymal transition of CRC cells. Taken together, the results of the current study revealed a novel molecular mechanism indicating that the miR‑151a‑5p/AGMAT axis could serve a crucial role in the regulation of CRC and could therefore be considered as a potential therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Yaya Xie
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Fengxian, Shanghai 201499, P.R. China
- School of Medical, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Yue Zhang
- School of Medical, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
- Hanzhong Central Hospital of Shaanxi, Hanzhong, Shaanxi 723000, P.R. China
| | - Xianju Liu
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Fengxian, Shanghai 201499, P.R. China
- School of Medical, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Lijun Cao
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Fengxian, Shanghai 201499, P.R. China
- School of Medical, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Mengting Han
- School of Medical, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Chunmei Wang
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Fengxian, Shanghai 201499, P.R. China
| | - Jinlian Chen
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Fengxian, Shanghai 201499, P.R. China
| | - Xingxing Zhang
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Fengxian, Shanghai 201499, P.R. China
| |
Collapse
|
6
|
Wilson LA, Pedroso MM, Peralta RA, Gahan LR, Schenk G. Biomimetics for purple acid phosphatases: A historical perspective. J Inorg Biochem 2023; 238:112061. [PMID: 36371912 DOI: 10.1016/j.jinorgbio.2022.112061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/23/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Biomimetics hold potential for varied applications in biotechnology and medicine but have also attracted particular interest as benchmarks for the functional study of their more complex biological counterparts, e.g. metalloenzymes. While many of the synthetic systems adequately mimic some structural and functional aspects of their biological counterparts the catalytic efficiencies displayed are mostly far inferior due to the smaller size and the associated lower complexity. Nonetheless they play an important role in bioinorganic chemistry. Numerous examples of biologically inspired and informed artificial catalysts have been reported, designed to mimic a plethora of chemical transformations, and relevant examples are highlighted in reviews and scientific reports. Herein, we discuss biomimetics of the metallohydrolase purple acid phosphatase (PAP), examples of which have been used to showcase synergistic research advances for both the biological and synthetic systems. In particular, we focus on the seminal contribution of our colleague Prof. Ademir Neves, and his group, pioneers in the design and optimization of suitable ligands that mimic the active site of PAP.
Collapse
Affiliation(s)
- Liam A Wilson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Marcelo M Pedroso
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Rosely A Peralta
- Departamento de Química, Universidade Federal de Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | - Lawrence R Gahan
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Gerhard Schenk
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia; Sustainable Minerals Institute, The University of Queensland, Brisbane, Queensland 4072, Australia; Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
7
|
Sinn M, Stanoppi M, Hauth F, Fleming JR, Funck D, Mayans O, Hartig JS. Guanidino acid hydrolysis by the human enzyme annotated as agmatinase. Sci Rep 2022; 12:22088. [PMID: 36543883 PMCID: PMC9772407 DOI: 10.1038/s41598-022-26655-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Guanidino acids such as taurocyamine, guanidinobutyrate, guanidinopropionate, and guanidinoacetate have been detected in humans. However, except for guanidionacetate, which is a precursor of creatine, their metabolism and potential functions remain poorly understood. Agmatine has received considerable attention as a potential neurotransmitter and the human enzyme so far annotated as agmatinase (AGMAT) has been proposed as an important modulator of agmatine levels. However, conclusive evidence for the assigned enzymatic activity is lacking. Here we show that AGMAT hydrolyzed a range of linear guanidino acids but was virtually inactive with agmatine. Structural modelling and direct biochemical assays indicated that two naturally occurring variants differ in their substrate preferences. A negatively charged group in the substrate at the end opposing the guanidine moiety was essential for efficient catalysis, explaining why agmatine was not hydrolyzed. We suggest to rename AGMAT as guanidino acid hydrolase (GDAH). Additionally, we demonstrate that the GDAH substrates taurocyamine, guanidinobutyrate and guanidinopropionate were produced by human glycine amidinotransferase (GATM). The presented findings show for the first time an enzymatic activity for GDAH/AGMAT. Since agmatine has frequently been proposed as an endogenous neurotransmitter, the current findings clarify important aspects of the metabolism of agmatine and guanidino acid derivatives in humans.
Collapse
Affiliation(s)
- Malte Sinn
- grid.9811.10000 0001 0658 7699Department of Chemistry, University of Konstanz, Konstanz, Germany
| | - Marco Stanoppi
- grid.9811.10000 0001 0658 7699Department of Chemistry, University of Konstanz, Konstanz, Germany
| | - Franziskus Hauth
- grid.9811.10000 0001 0658 7699Department of Chemistry, University of Konstanz, Konstanz, Germany ,grid.9811.10000 0001 0658 7699Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Konstanz, Germany
| | - Jennifer R. Fleming
- grid.9811.10000 0001 0658 7699Department of Biology, University of Konstanz, Konstanz, Germany
| | - Dietmar Funck
- grid.9811.10000 0001 0658 7699Department of Chemistry, University of Konstanz, Konstanz, Germany
| | - Olga Mayans
- grid.9811.10000 0001 0658 7699Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Konstanz, Germany ,grid.9811.10000 0001 0658 7699Department of Biology, University of Konstanz, Konstanz, Germany
| | - Jörg S. Hartig
- grid.9811.10000 0001 0658 7699Department of Chemistry, University of Konstanz, Konstanz, Germany ,grid.9811.10000 0001 0658 7699Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Konstanz, Germany
| |
Collapse
|
8
|
New Insights into the Determinants of Specificity in Human Type I Arginase: Generation of a Mutant That Is Only Active with Agmatine as Substrate. Int J Mol Sci 2022; 23:ijms23126438. [PMID: 35742891 PMCID: PMC9224512 DOI: 10.3390/ijms23126438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/25/2022] [Accepted: 06/04/2022] [Indexed: 02/04/2023] Open
Abstract
Arginase catalyzes the hydrolysis of L-arginine into L-ornithine and urea. This enzyme has several analogies with agmatinase, which catalyzes the hydrolysis of agmatine into putrescine and urea. However, this contrasts with the highlighted specificity that each one presents for their respective substrate. A comparison of available crystal structures for arginases reveals an important difference in the extension of two loops located in the entrance of the active site. The first, denominated loop A (I129-L140) contains the residues that interact with the alpha carboxyl group or arginine of arginase, and the loop B (D181-P184) contains the residues that interact with the alpha amino group of arginine. In this work, to determine the importance of these loops in the specificity of arginase, single, double, and triple arginase mutants in these loops were constructed, as well as chimeras between type I human arginase and E. coli agmatinase. In previous studies, the substitution of N130D in arginase (in loop A) generated a species capable of hydrolyzing arginine and agmatine. Now, the specificity of arginase is completely altered, generating a chimeric species that is only active with agmatine as a substrate, by substituting I129T, N130Y, and T131A together with the elimination of residues P132, L133, and T134. In addition, Quantum Mechanic/Molecular Mechanic (QM/MM) calculations were carried out to study the accommodation of the substrates in in the active site of this chimera. With these results it is concluded that this loop is decisive to discriminate the type of substrate susceptible to be hydrolyzed by arginase. Evidence was also obtained to define the loop B as a structural determinant for substrate affinity. Concretely, the double mutation D181T and V182E generate an enzyme with an essentially unaltered kcat value, but with a significantly increased Km value for arginine and a significant decrease in affinity for its product ornithine.
Collapse
|
9
|
Zhang Y, Cao L, Xie Y, Wang C, Liu X, Zhang X, Chen J. Agmatinase facilitates the tumorigenesis of pancreatic adenocarcinoma through the TGFβ/Smad pathway. Exp Ther Med 2022; 24:490. [PMID: 35837051 PMCID: PMC9257765 DOI: 10.3892/etm.2022.11417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/19/2022] [Indexed: 11/08/2022] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is one of the most lethal malignancies. Due to the lack of typical symptoms and difficulties in early diagnosis, PAAD has a high mortality rate. Therefore, it is essential to identify novel specific biomarkers for the application of targeted therapies. A previous study suggested that agmatinase (AGMAT) may fulfill important roles in tumor progression; however, these roles and the underlying mechanisms of AGMAT involvement in PAAD have yet to be thoroughly investigated. To address this shortcoming, in the present study the expression and prognostic significance of AGMAT were analyzed via several bioinformatics databases. Gain- and loss-of-function experiments were subsequently performed to observe the impact of AGMAT on the proliferation and metastasis of PAAD cells via Cell Counting Kit 8 (CCK-8) assay, colony formation assay, and cell migration and invasion assays in vitro. In order to probe the mechanisms involved, western blot assays were performed. AGMAT was found to be overexpressed in PAAD, and it was positively associated with a poor prognosis. Stable overexpression of AGMAT was found to lead to a marked increase in cell proliferation and metastasis through activation of the transforming growth factor-β (TGFβ)/Smad pathway, and via enhancing epithelial-mesenchymal transition (EMT). In conclusion, the results of the present study suggest that AGMAT may be an oncogene, and a pivotal mechanism has been uncovered in which AGMAT facilitates the progression of PAAD tumorigenesis through the TGFβ/Smad pathway.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai 201499, P.R. China
| | - Lijun Cao
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai 201499, P.R. China
| | - Yaya Xie
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai 201499, P.R. China
| | - Chunmei Wang
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai 201499, P.R. China
| | - Xianju Liu
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai 201499, P.R. China
| | - Xingxing Zhang
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai 201499, P.R. China
| | - Jinlian Chen
- Department of Gastroenterology, Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai 201499, P.R. China
| |
Collapse
|
10
|
Funck D, Sinn M, Fleming JR, Stanoppi M, Dietrich J, López-Igual R, Mayans O, Hartig JS. Discovery of a Ni 2+-dependent guanidine hydrolase in bacteria. Nature 2022; 603:515-521. [PMID: 35264792 DOI: 10.1038/s41586-022-04490-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 01/31/2022] [Indexed: 12/14/2022]
Abstract
Nitrogen availability is a growth-limiting factor in many habitats1, and the global nitrogen cycle involves prokaryotes and eukaryotes competing for this precious resource. Only some bacteria and archaea can fix elementary nitrogen; all other organisms depend on the assimilation of mineral or organic nitrogen. The nitrogen-rich compound guanidine occurs widely in nature2-4, but its utilization is impeded by pronounced resonance stabilization5, and enzymes catalysing hydrolysis of free guanidine have not been identified. Here we describe the arginase family protein GdmH (Sll1077) from Synechocystis sp. PCC 6803 as a Ni2+-dependent guanidine hydrolase. GdmH is highly specific for free guanidine. Its activity depends on two accessory proteins that load Ni2+ instead of the typical Mn2+ ions into the active site. Crystal structures of GdmH show coordination of the dinuclear metal cluster in a geometry typical for arginase family enzymes and allow modelling of the bound substrate. A unique amino-terminal extension and a tryptophan residue narrow the substrate-binding pocket and identify homologous proteins in further cyanobacteria, several other bacterial taxa and heterokont algae as probable guanidine hydrolases. This broad distribution suggests notable ecological relevance of guanidine hydrolysis in aquatic habitats.
Collapse
Affiliation(s)
- D Funck
- Department of Chemistry, University of Konstanz, Konstanz, Germany
| | - M Sinn
- Department of Chemistry, University of Konstanz, Konstanz, Germany
| | - J R Fleming
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - M Stanoppi
- Department of Chemistry, University of Konstanz, Konstanz, Germany
| | - J Dietrich
- Department of Chemistry, University of Konstanz, Konstanz, Germany
| | - R López-Igual
- Instituto de Bioquímica Vegetal y Fotosíntesis, Universidad de Sevilla and C.S.I.C, Seville, Spain
| | - O Mayans
- Department of Biology, University of Konstanz, Konstanz, Germany.,Konstanz Graduate School Chemical Biology (KoRS-CB), University of Konstanz, Konstanz, Germany
| | - J S Hartig
- Department of Chemistry, University of Konstanz, Konstanz, Germany. .,Konstanz Graduate School Chemical Biology (KoRS-CB), University of Konstanz, Konstanz, Germany.
| |
Collapse
|
11
|
Muller J, Attia R, Zedet A, Girard C, Pudlo M. An Update on Arginase Inhibitors and Inhibitory Assays. Mini Rev Med Chem 2021; 22:1963-1976. [PMID: 34967285 DOI: 10.2174/1389557522666211229105703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/18/2021] [Accepted: 10/22/2021] [Indexed: 11/22/2022]
Abstract
Arginase, which converts arginine into ornithine and urea, is a promising therapeutic target. Arginase is involved in cardiovascular diseases, parasitic infections and, through a critical role in immunity, in some cancers. There is a need to develop effective arginase inhibitors and therefore efforts to identify and optimize new inhibitors are increasing. Several methods of evaluating arginase activity are available, but few directly measure the product. Radiometric assays need to separate urea and dying reactions require acidic conditions and sometimes heating. Hence, there are a variety of different approaches available, and each approach has its own limits and benefits. In this review, we provide an update on arginase inhibitors, followed by a discussion on available arginase assays and alternative methods, with a focus on the intrinsic biases and parameters that are likely to impact results.
Collapse
Affiliation(s)
- Jason Muller
- PEPITE EA4267, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| | - Rym Attia
- PEPITE EA4267, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| | - Andy Zedet
- PEPITE EA4267, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| | - Corine Girard
- PEPITE EA4267, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| | - Marc Pudlo
- PEPITE EA4267, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| |
Collapse
|
12
|
Pérez-Mozqueda LL, Vazquez-Duhalt R, Castro-Longoria E. Enzymatic characterization of agmatinase (AGM-1) from the filamentous fungus Neurospora crassa. Fungal Genet Biol 2021; 157:103634. [PMID: 34634482 DOI: 10.1016/j.fgb.2021.103634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 10/20/2022]
Abstract
Agmatinase is a metallohydrolase involved in the hydrolysis of agmatine to produce urea and putrescine. Although its role in organisms is still under study, there are no reports of this family of enzymes in filamentous fungi. Recently, a protein showing agmatinase activity was reported in Neurospora crassa. Therefore, the aim of this work is to determine if the protein (AGM-1) found in the filamentous fungus N. crassa is a true agmatinase. The protein AGM-1was purified directly from N. crassa cultures, and its enzymatic characterization was carried out. The catalytic parameters such as optimum pH, thermostability, transformation kinetics, and activity in the presence of a cofactor were determined. The results show that AGM-1 can use manganese as a cofactor for its enzymatic activity, showing a transformation rate constant (kcat) of 77 s-1 and an affinity constant (KM) of 50.5 mM. The protein loses 50% of its activity when incubated 15 min at 30 °C and reaches maximal enzymatic activity at a pH range of 8-8.5. Our results indicate that the AGM-1 from N. crassa shows similar characteristics to true agmatinases already reported in other organisms. Thus, our findings strongly support that the protein annotated as hypothetical agmatinase in N. crassa is a true agmatinase.
Collapse
Affiliation(s)
- Luis L Pérez-Mozqueda
- Departamento de Microbiología, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada, B.C, Mexico
| | - Rafael Vazquez-Duhalt
- Centro de Nanociencias y Nanotecnología (CNyN), Universidad Nacional Autónoma de México (UNAM), Ensenada, B.C, Mexico
| | - Ernestina Castro-Longoria
- Departamento de Microbiología, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada, B.C, Mexico.
| |
Collapse
|
13
|
Feder D, Mohd-Pahmi SH, Hussein WM, Guddat LW, McGeary RP, Schenk G. Rational Design of Potent Inhibitors of a Metallohydrolase Using a Fragment-Based Approach. ChemMedChem 2021; 16:3342-3359. [PMID: 34331400 DOI: 10.1002/cmdc.202100486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Indexed: 11/08/2022]
Abstract
Metallohydrolases form a large group of enzymes that have fundamental importance in a broad range of biological functions. Among them, the purple acid phosphatases (PAPs) have gained attention due to their crucial role in the acquisition and use of phosphate by plants and also as a promising target for novel treatments of bone-related disorders and cancer. To date, no crystal structure of a mammalian PAP with drug-like molecules bound near the active site is available. Herein, we used a fragment-based design approach using structures of a mammalian PAP in complex with the MaybridgeTM fragment CC063346, the amino acid L-glutamine and the buffer molecule HEPES, as well as various solvent molecules to guide the design of highly potent and efficient mammalian PAP inhibitors. These inhibitors have improved aqueous solubility when compared to the clinically most promising PAP inhibitors available to date. Furthermore, drug-like fragments bound in newly discovered binding sites mapped out additional scaffolds for further inhibitor discovery, as well as scaffolds for the design of inhibitors with novel modes of action.
Collapse
Affiliation(s)
- Daniel Feder
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia.,Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Siti H Mohd-Pahmi
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Waleed M Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Luke W Guddat
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Ross P McGeary
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Gerhard Schenk
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia.,Sustainable Minerals Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
14
|
Hernández VM, Arteaga A, Dunn MF. Diversity, properties and functions of bacterial arginases. FEMS Microbiol Rev 2021; 45:6308370. [PMID: 34160574 DOI: 10.1093/femsre/fuab034] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/17/2021] [Indexed: 02/07/2023] Open
Abstract
The metalloenzyme arginase hydrolyzes L-arginine to produce L-ornithine and urea. In bacteria, arginase has important functions in basic nitrogen metabolism and redistribution, production of the key metabolic precursor L-ornithine, stress resistance and pathogenesis. We describe the regulation and specific functions of the arginase pathway as well as summarize key characteristics of related arginine catabolic pathways. The use of arginase-derived ornithine as a precursor molecule is reviewed. We discuss the biochemical and transcriptional regulation of arginine metabolism, including arginase, with the latter topic focusing on the RocR and AhrC transcriptional regulators in the model organism Bacillus subtilis. Finally, we consider similarities and contrasts in the structure and catalytic mechanism of the arginases from Bacillus caldovelox and Helicobacter pylori. The overall aim of this review is to provide a panorama of the diversity of physiological functions, regulation, and biochemical features of arginases in a variety of bacterial species.
Collapse
Affiliation(s)
- Victor M Hernández
- Programa de Genómica Funcional de Procariotes, Centro de Ciencias Genómicas-Universidad Nacional Autonoma de México, Cuernavaca, Morelos, C.P. 62210, Mexico
| | - Alejandra Arteaga
- Programa de Genómica Funcional de Procariotes, Centro de Ciencias Genómicas-Universidad Nacional Autonoma de México, Cuernavaca, Morelos, C.P. 62210, Mexico
| | - Michael F Dunn
- Programa de Genómica Funcional de Procariotes, Centro de Ciencias Genómicas-Universidad Nacional Autonoma de México, Cuernavaca, Morelos, C.P. 62210, Mexico
| |
Collapse
|
15
|
Maturana P, Orellana MS, Herrera SM, Martínez I, Figueroa M, Martínez-Oyanedel J, Castro-Fernandez V, Uribe E. Crystal Structure of Escherichia coli Agmatinase: Catalytic Mechanism and Residues Relevant for Substrate Specificity. Int J Mol Sci 2021; 22:ijms22094769. [PMID: 33946272 PMCID: PMC8125230 DOI: 10.3390/ijms22094769] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/18/2021] [Accepted: 04/28/2021] [Indexed: 11/16/2022] Open
Abstract
Agmatine is the product of the decarboxylation of L-arginine by the enzyme arginine decarboxylase. This amine has been attributed to neurotransmitter functions, anticonvulsant, anti-neurotoxic, and antidepressant in mammals and is a potential therapeutic agent for diseases such as Alzheimer's, Parkinson's, and cancer. Agmatinase enzyme hydrolyze agmatine into urea and putrescine, which belong to one of the pathways producing polyamines, essential for cell proliferation. Agmatinase from Escherichia coli (EcAGM) has been widely studied and kinetically characterized, described as highly specific for agmatine. In this study, we analyze the amino acids involved in the high specificity of EcAGM, performing a series of mutations in two loops critical to the active-site entrance. Two structures in different space groups were solved by X-ray crystallography, one at low resolution (3.2 Å), including a guanidine group; and other at high resolution (1.8 Å) which presents urea and agmatine in the active site. These structures made it possible to understand the interface interactions between subunits that allow the hexameric state and postulate a catalytic mechanism according to the Mn2+ and urea/guanidine binding site. Molecular dynamics simulations evaluated the conformational dynamics of EcAGM and residues participating in non-binding interactions. Simulations showed the high dynamics of loops of the active site entrance and evidenced the relevance of Trp68, located in the adjacent subunit, to stabilize the amino group of agmatine by cation-pi interaction. These results allow to have a structural view of the best-kinetic characterized agmatinase in literature up to now.
Collapse
Affiliation(s)
- Pablo Maturana
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Ñuñoa 7800003, Santiago, Chile; (P.M.); (S.M.H.)
| | - María S. Orellana
- Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago 8370251, Santiago, Chile;
| | - Sixto M. Herrera
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Ñuñoa 7800003, Santiago, Chile; (P.M.); (S.M.H.)
| | - Ignacio Martínez
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción 4070386, Concepción, Chile; (I.M.); (M.F.); (J.M.-O.)
| | - Maximiliano Figueroa
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción 4070386, Concepción, Chile; (I.M.); (M.F.); (J.M.-O.)
| | - José Martínez-Oyanedel
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción 4070386, Concepción, Chile; (I.M.); (M.F.); (J.M.-O.)
| | - Victor Castro-Fernandez
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Ñuñoa 7800003, Santiago, Chile; (P.M.); (S.M.H.)
- Correspondence: (V.C.-F.); (E.U.); Tel.: +56-2-2978-7332 (V.C.-F.); +56-41-220-4428 (E.U.)
| | - Elena Uribe
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción 4070386, Concepción, Chile; (I.M.); (M.F.); (J.M.-O.)
- Correspondence: (V.C.-F.); (E.U.); Tel.: +56-2-2978-7332 (V.C.-F.); +56-41-220-4428 (E.U.)
| |
Collapse
|
16
|
Tinkov AA, Paoliello MMB, Mazilina AN, Skalny AV, Martins AC, Voskresenskaya ON, Aaseth J, Santamaria A, Notova SV, Tsatsakis A, Lee E, Bowman AB, Aschner M. Molecular Targets of Manganese-Induced Neurotoxicity: A Five-Year Update. Int J Mol Sci 2021; 22:4646. [PMID: 33925013 PMCID: PMC8124173 DOI: 10.3390/ijms22094646] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/22/2021] [Accepted: 04/25/2021] [Indexed: 12/16/2022] Open
Abstract
Understanding of the immediate mechanisms of Mn-induced neurotoxicity is rapidly evolving. We seek to provide a summary of recent findings in the field, with an emphasis to clarify existing gaps and future research directions. We provide, here, a brief review of pertinent discoveries related to Mn-induced neurotoxicity research from the last five years. Significant progress was achieved in understanding the role of Mn transporters, such as SLC39A14, SLC39A8, and SLC30A10, in the regulation of systemic and brain manganese handling. Genetic analysis identified multiple metabolic pathways that could be considered as Mn neurotoxicity targets, including oxidative stress, endoplasmic reticulum stress, apoptosis, neuroinflammation, cell signaling pathways, and interference with neurotransmitter metabolism, to name a few. Recent findings have also demonstrated the impact of Mn exposure on transcriptional regulation of these pathways. There is a significant role of autophagy as a protective mechanism against cytotoxic Mn neurotoxicity, yet also a role for Mn to induce autophagic flux itself and autophagic dysfunction under conditions of decreased Mn bioavailability. This ambivalent role may be at the crossroad of mitochondrial dysfunction, endoplasmic reticulum stress, and apoptosis. Yet very recent evidence suggests Mn can have toxic impacts below the no observed adverse effect of Mn-induced mitochondrial dysfunction. The impact of Mn exposure on supramolecular complexes SNARE and NLRP3 inflammasome greatly contributes to Mn-induced synaptic dysfunction and neuroinflammation, respectively. The aforementioned effects might be at least partially mediated by the impact of Mn on α-synuclein accumulation. In addition to Mn-induced synaptic dysfunction, impaired neurotransmission is shown to be mediated by the effects of Mn on neurotransmitter systems and their complex interplay. Although multiple novel mechanisms have been highlighted, additional studies are required to identify the critical targets of Mn-induced neurotoxicity.
Collapse
Affiliation(s)
- Alexey A. Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia;
- Laboratory of Molecular Dietetics, Department of Neurological Diseases and Neurosurgery, Department of Analytical and Forensic Toxicology, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (O.N.V.); (J.A.); (A.T.)
| | - Monica M. B. Paoliello
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.M.B.P.); (A.C.M.)
- Graduate Program in Public Health, Center of Health Sciences, State University of Londrina, Londrina, PR 86038-350, Brazil
| | - Aksana N. Mazilina
- Department of Medical Elementology, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia;
| | - Anatoly V. Skalny
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia;
- Laboratory of Medical Elementology, KG Razumovsky Moscow State University of Technologies and Management, 109004 Moscow, Russia
| | - Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.M.B.P.); (A.C.M.)
| | - Olga N. Voskresenskaya
- Laboratory of Molecular Dietetics, Department of Neurological Diseases and Neurosurgery, Department of Analytical and Forensic Toxicology, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (O.N.V.); (J.A.); (A.T.)
| | - Jan Aaseth
- Laboratory of Molecular Dietetics, Department of Neurological Diseases and Neurosurgery, Department of Analytical and Forensic Toxicology, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (O.N.V.); (J.A.); (A.T.)
- Research Department, Innlandet Hospital Trust, P.O. Box 104, 2381 Brumunddal, Norway
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, SSA, Mexico City 14269, Mexico;
| | - Svetlana V. Notova
- Institute of Bioelementology, Orenburg State University, 460018 Orenburg, Russia;
- Federal Research Centre of Biological Systems and Agro-technologies of the Russian Academy of Sciences, 460000 Orenburg, Russia
| | - Aristides Tsatsakis
- Laboratory of Molecular Dietetics, Department of Neurological Diseases and Neurosurgery, Department of Analytical and Forensic Toxicology, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (O.N.V.); (J.A.); (A.T.)
- Laboratory of Toxicology, Medical School, University of Crete, Voutes, 700 13 Heraklion, Greece
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47906, USA;
| | - Michael Aschner
- Laboratory of Molecular Dietetics, Department of Neurological Diseases and Neurosurgery, Department of Analytical and Forensic Toxicology, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (O.N.V.); (J.A.); (A.T.)
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.M.B.P.); (A.C.M.)
| |
Collapse
|
17
|
Chitrakar I, Ahmed SF, Torelli AT, French JB. Structure of the E. coli agmatinase, SPEB. PLoS One 2021; 16:e0248991. [PMID: 33857156 PMCID: PMC8049259 DOI: 10.1371/journal.pone.0248991] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/09/2021] [Indexed: 01/05/2023] Open
Abstract
Agmatine amidinohydrolase, or agmatinase, catalyzes the conversion of agmatine to putrescine and urea. This enzyme is found broadly across kingdoms of life and plays a critical role in polyamine biosynthesis and the regulation of agmatine concentrations. Here we describe the high-resolution X-ray crystal structure of the E. coli agmatinase, SPEB. The data showed a relatively high degree of pseudomerohedral twinning, was ultimately indexed in the P31 space group and led to a final model with eighteen chains, corresponding to three full hexamers in the asymmetric unit. There was a solvent content of 38.5% and refined R/Rfree values of 0.166/0.216. The protein has the conserved fold characteristic of the agmatine ureohydrolase family and displayed a high degree of structural similarity among individual protomers. Two distinct peaks of electron density were observed in the active site of most of the eighteen chains of SPEB. As the activity of this protein is known to be dependent upon manganese and the fold is similar to other dinuclear metallohydrolases, these peaks were modeled as manganese ions. The orientation of the conserved active site residues, in particular those amino acids that participate in binding the metal ions and a pair of acidic residues (D153 and E274 in SPEB) that play a role in catalysis, are similar to other agmatinase and arginase enzymes and is consistent with a hydrolytic mechanism that proceeds via a metal-activated hydroxide ion.
Collapse
Affiliation(s)
- Iva Chitrakar
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, United States of America
- Biochemistry and Structural Biology Graduate Program, Stony Brook University, Stony Brook, NY, United States of America
| | - Syed Fardin Ahmed
- Department of Chemistry, Ithaca College, Ithaca, NY, United States of America
| | - Andrew T. Torelli
- Department of Chemistry, Ithaca College, Ithaca, NY, United States of America
| | - Jarrod B. French
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, United States of America
- Chemistry Department, Stony Brook University, Stony Brook, NY, United States of America
- Hormel Institute, University of Minnesota, Austin, MN, United States of America
| |
Collapse
|
18
|
Zhao J, Wang L, Wei X, Li K, Liu J. Food-Grade Expression and Characterization of a Dextranase from Chaetomium gracile Suitable for Sugarcane Juice Clarification. Chem Biodivers 2020; 18:e2000797. [PMID: 33245200 DOI: 10.1002/cbdv.202000797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/26/2020] [Indexed: 11/09/2022]
Abstract
The microbial production of dextranase using cheap carbon sources is beneficial to solve the economic loss caused by the accumulation of dextran in syrup. A food-grade microbial cell factory was constructed by introducing the dextranase encoding gene DEX from Chaetomium gracile to the chromosome of Bacillus subtilis, and the antibiotic resistance marker gene was subsequently deleted via the Cre/loxP strategy. The dual-promoter system with a sequentially arranged constitutive P43 promoter resulted in an 85 % increase in DEX expression. Under the optimal fermentation conditions of 10 g/L maltose, 15 g/L casein, 1 g/L Na2 HPO4 , 1 g/L FeSO4 and 8 g/L NaCl, DEX activity was increased from 2.625 to 64.34 U/mL. Recombinant DEX was purified 5.98-fold with a recovery ratio of 26.67 % and specific activity of 3935.02 U/mg. Enzyme activity was optimal at 55 °C and pH 5.0 and remained 80.34 % and 71.36 % of the initial activity at 55 °C and pH 4.0 after 60 min, respectively. The enzyme possessed high activity in the presence of Co2+ , while Ag+ showed the strongest inhibition ability. The optimal substrate was 20 g/L dextran T-2000. The findings could facilitate the low-cost, large-scale production of food-grade DEX for use in the sugar industry.
Collapse
Affiliation(s)
- Jingyi Zhao
- College of Light Industry and Food Engineering, Guangxi University, 100 Daxue Road, Nanning, Guangxi, 530004, P. R. China
| | - Leyi Wang
- College of Light Industry and Food Engineering, Guangxi University, 100 Daxue Road, Nanning, Guangxi, 530004, P. R. China
| | - Xin Wei
- College of Light Industry and Food Engineering, Guangxi University, 100 Daxue Road, Nanning, Guangxi, 530004, P. R. China
| | - Kai Li
- College of Light Industry and Food Engineering, Guangxi University, 100 Daxue Road, Nanning, Guangxi, 530004, P. R. China.,Sugar Industry Collaborative Innovation Center, Guangxi University, 100 Daxue Road, Nanning, Guangxi, 530004, P. R. China
| | - Jidong Liu
- College of Light Industry and Food Engineering, Guangxi University, 100 Daxue Road, Nanning, Guangxi, 530004, P. R. China.,Sugar Industry Collaborative Innovation Center, Guangxi University, 100 Daxue Road, Nanning, Guangxi, 530004, P. R. China
| |
Collapse
|
19
|
The structure-based reaction mechanism of urease, a nickel dependent enzyme: tale of a long debate. J Biol Inorg Chem 2020; 25:829-845. [PMID: 32809087 PMCID: PMC7433671 DOI: 10.1007/s00775-020-01808-w] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 06/29/2020] [Indexed: 01/22/2023]
Abstract
This review is an attempt to retrace the chronicle that starts from the discovery of the role of nickel as the essential metal ion in urease for the enzymatic catalysis of urea, a key step in the biogeochemical cycle of nitrogen on Earth, to the most recent progress in understanding the chemistry of this historical enzyme. Data and facts are presented through the magnifying lenses of the authors, using their best judgment to filter and elaborate on the many facets of the research carried out on this metalloenzyme over the years. The tale is divided in chapters that discuss and describe the results obtained in the subsequent leaps in the knowledge that led from the discovery of a biological role for Ni to the most recent advancements in the comprehension of the relationship between the structure and function of urease. This review is intended not only to focus on the bioinorganic chemistry of this beautiful metal-based catalysis, but also, and maybe primarily, to evoke inspiration and motivation to further explore the realm of bio-based coordination chemistry.
Collapse
|
20
|
Finke H, Winkelbeiner N, Lossow K, Hertel B, Wandt VK, Schwarz M, Pohl G, Kopp JF, Ebert F, Kipp AP, Schwerdtle T. Effects of a Cumulative, Suboptimal Supply of Multiple Trace Elements in Mice: Trace Element Status, Genomic Stability, Inflammation, and Epigenetics. Mol Nutr Food Res 2020; 64:e2000325. [PMID: 32609929 DOI: 10.1002/mnfr.202000325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/05/2020] [Indexed: 12/15/2022]
Abstract
SCOPE Trace element (TE) deficiencies often occur accumulated, as nutritional intake is inadequate for several TEs, concurrently. Therefore, the impact of a suboptimal supply of iron, zinc, copper, iodine, and selenium on the TE status, health parameters, epigenetics, and genomic stability in mice are studied. METHODS AND RESULTS Male mice receive reduced or adequate amounts of TEs for 9 weeks. The TE status is analyzed mass-spectrometrically in serum and different tissues. Furthermore, gene and protein expression of TE biomarkers are assessed with focus on liver. Iron concentrations are most sensitive toward a reduced supply indicated by increased serum transferrin levels and altered hepatic expression of iron-related genes. Reduced TE supply results in smaller weight gain but higher spleen and heart weights. Additionally, inflammatory mediators in serum and liver are increased together with hepatic genomic instability. However, global DNA (hydroxy)methylation is unaffected by the TE modulation. CONCLUSION Despite homeostatic regulation of most TEs in response to a low intake, this condition still has substantial effects on health parameters. It appears that the liver and immune system react particularly sensitive toward changes in TE intake. The reduced Fe status might be the primary driver for the observed effects.
Collapse
Affiliation(s)
- Hannah Finke
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany
| | - Nicola Winkelbeiner
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Kristina Lossow
- Department of Molecular Nutritional Physiology, Institute of Nutritional Sciences, Friedrich-Schiller University Jena, Dornburger Straße 24, Jena, 07743, Germany.,German Institute of Human Nutrition, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Barbara Hertel
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany
| | - Viktoria K Wandt
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Maria Schwarz
- Department of Molecular Nutritional Physiology, Institute of Nutritional Sciences, Friedrich-Schiller University Jena, Dornburger Straße 24, Jena, 07743, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Gabriele Pohl
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany
| | - Johannes F Kopp
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Franziska Ebert
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Anna P Kipp
- Department of Molecular Nutritional Physiology, Institute of Nutritional Sciences, Friedrich-Schiller University Jena, Dornburger Straße 24, Jena, 07743, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Tanja Schwerdtle
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany.,German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, Berlin, 10589, Germany
| |
Collapse
|
21
|
Insights into the Mn 2+ Binding Site in the Agmatinase-Like Protein (ALP): A Critical Enzyme for the Regulation of Agmatine Levels in Mammals. Int J Mol Sci 2020; 21:ijms21114132. [PMID: 32531922 PMCID: PMC7313459 DOI: 10.3390/ijms21114132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 11/21/2022] Open
Abstract
Agmatine is a neurotransmitter with anticonvulsant, anti-neurotoxic and antidepressant-like effects, in addition it has hypoglycemic actions. Agmatine is converted to putrescine and urea by agmatinase (AGM) and by an agmatinase-like protein (ALP), a new type of enzyme which is present in human and rodent brain tissues. Recombinant rat brain ALP is the only mammalian protein that exhibits significant agmatinase activity in vitro and generates putrescine under in vivo conditions. ALP, despite differing in amino acid sequence from all members of the ureohydrolase family, is strictly dependent on Mn2+ for catalytic activity. However, the Mn2+ ligands have not yet been identified due to the lack of structural information coupled with the low sequence identity that ALPs display with known ureohydrolases. In this work, we generated a structural model of the Mn2+ binding site of the ALP and we propose new putative Mn2+ ligands. Then, we cloned and expressed a sequence of 210 amino acids, here called the “central-ALP”, which include the putative ligands of Mn2+. The results suggest that the central-ALP is catalytically active, as agmatinase, with an unaltered Km for agmatine and a decreased kcat. Similar to wild-type ALP, central-ALP is activated by Mn2+ with a similar affinity. Besides, a simple mutant D217A, a double mutant E288A/K290A, and a triple mutant N213A/Q215A/D217A of these putative Mn2+ ligands result on the loss of ALP agmatinase activity. Our results indicate that the central-ALP contains the active site for agmatine hydrolysis, as well as that the residues identified are relevant for the ALP catalysis.
Collapse
|
22
|
Lobos M, Figueroa M, Martínez-Oyanedel J, López V, García-Robles MDLÁ, Tarifeño-Saldivia E, Carvajal N, Uribe E. Insights on the participation of Glu256 and Asp204 in the oligomeric structure and cooperative effects of human arginase type I. J Struct Biol 2020; 211:107533. [PMID: 32450233 DOI: 10.1016/j.jsb.2020.107533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 11/20/2022]
Abstract
Arginase (EC 3.5.3.1) catalyzes the hydrolysis of L-arginine to L-ornithine and urea, and requires a bivalent cation, especially Mn2+ for its catalytic activity. It is a component of the urea cycle and regulates the intracellular levels of l-arginine, which makes the arginase a target for treatment of vascular diseases and asthma. Mammalian arginases contain an unusual S-shaped motif located at the intermonomeric interface. Until now, the studies were limited to structural role of the motif. Then, our interest was focused on functional aspects and our hypothesis has been that the motif is essential for maintain the oligomeric state, having Arg308 as a central axis. Previously, we have shown that the R308A mutant is monomeric and re-associates to the trimeric-cooperative state in the presence of low concentrations of guanidine chloride. We have now mutated Asp204 that interacts with Arg308 in the neighbor subunit, and also we mutated Glu256, proposed as important for oligomerization. Concretely, the human arginase I mutants D204A, D204E, E256A, E256Q and E256D were generated and examined. No differences were observed in the kinetic parameters at pH 9.5 or in tryptophan fluorescence. However, the D204A and E256Q variants were monomeric. On the other hand, D204E and E256D proved to be trimeric and kinetically cooperative at pH 7.5, whereas hyperbolic kinetics was exhibited by E256A, also trimeric. The results obtained strongly support the importance of the interaction between Arg255 and Glu256 in the cooperative properties of arginase, and Asp204 would be relevant to maintain the oligomeric state through salt bridges with Arg255 and Arg308.
Collapse
Affiliation(s)
- Marcela Lobos
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - Maximiliano Figueroa
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción, Chile.
| | - José Martínez-Oyanedel
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - Vasthi López
- Departamento de Ciencias Biomédicas. Universidad Católica del Norte, Coquimbo, Chile
| | | | - Estefanía Tarifeño-Saldivia
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - Nelson Carvajal
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - Elena Uribe
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción, Chile.
| |
Collapse
|
23
|
Ide AA, Hernández VM, Medina-Aparicio L, Carcamo-Noriega E, Girard L, Hernández-Lucas I, Dunn MF. Genetic regulation, biochemical properties and physiological importance of arginase from Sinorhizobium meliloti. Microbiology (Reading) 2020; 166:484-497. [DOI: 10.1099/mic.0.000909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In bacteria,l-arginine is a precursor of various metabolites and can serve as a source of carbon and/or nitrogen. Arginine catabolism by arginase, which hydrolyzes arginine tol-ornithine and urea, is common in nature but has not been studied in symbiotic nitrogen-fixing rhizobia. The genome of the alfalfa microsymbiontSinorhizobium meliloti1021 has two genes annotated as arginases,argI1(smc03091) andargI2(sma1711). Biochemical assays with purified ArgI1 and ArgI2 (as 6His-Sumo-tagged proteins) showed that only ArgI1 had detectable arginase activity. A 1021argI1null mutant lacked arginase activity and grew at a drastically reduced rate with arginine as sole nitrogen source. Wild-type growth and arginase activity were restored in theargI1mutant genetically complemented with a genomically integratedargI1gene. In the wild-type, arginase activity andargI1transcription were induced several fold by exogenous arginine. ArgI1 purified as a 6His-Sumo-tagged protein had its highestin vitroenzymatic activity at pH 7.5 with Ni2+as cofactor. The enzyme was also active with Mn2+and Co2+, both of which gave the enzyme the highest activities at a more alkaline pH. The 6His-Sumo-ArgI1 comprised three identical subunits based on the migration of the urea-dissociated protein in a native polyacrylamide gel. A Lrp-like regulator (smc03092) divergently transcribed fromargI1was required for arginase induction by arginine or ornithine. This regulator was designated ArgIR. Electrophoretic mobility shift assays showed that purified ArgIR bound to theargI1promoter in a region preceding the predictedargI1transcriptional start. Our results indicate that ArgI1 is the sole arginase inS. meliloti, that it contributes substantially to arginine catabolismin vivoand thatargI1induction by arginine is dependent on ArgIR.
Collapse
Affiliation(s)
- Alejandra Arteaga Ide
- Programa de Genómica Funcional de Procariotes, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| | - Victor M. Hernández
- Programa de Genómica Funcional de Procariotes, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| | - Liliana Medina-Aparicio
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| | - Edson Carcamo-Noriega
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| | - Lourdes Girard
- Programa de Biología de Sistemas y Biología Sintética, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| | - Ismael Hernández-Lucas
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| | - Michael F. Dunn
- Programa de Genómica Funcional de Procariotes, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| |
Collapse
|