51
|
Costa TR, Amstalden MK, Ribeiro DL, Menaldo DL, Sartim MA, Aissa AF, Antunes LMG, Sampaio SV. CR-LAAO causes genotoxic damage in HepG2 tumor cells by oxidative stress. Toxicology 2018; 404-405:42-48. [PMID: 29738841 DOI: 10.1016/j.tox.2018.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/23/2018] [Accepted: 05/04/2018] [Indexed: 12/28/2022]
Abstract
Snake venom L-amino acid oxidases (SV-LAAOs) are enzymes of great interest in research due to their many biological effects with therapeutic potential. CR-LAAO, an L-amino acid oxidase from Calloselasma rhodostoma snake venom, is a well described SV-LAAO with immunomodulatory, antiparasitic, microbicidal, and antitumor effects. In this study, we evaluated the genotoxic potential of this enzyme in human peripheral blood mononuclear cells (PBMC) and HepG2 tumor cells, as well as its interaction with these cells, its impact on the expression of DNA repair and antioxidant pathway genes, and reactive oxygen species (ROS)-induced intracellular production. Flow cytometry analysis of FITC-labelled CR-LAAO showed higher specificity of interaction with HepG2 cells than PBMC. Moreover, CR-LAAO significantly increased intracellular levels of ROS only in HepG2 tumor cells, as assessed by fluorescence. CR-LAAO also induced genotoxicity in HepG2 cells and PBMC after 4 h of stimulus, with DNA damages persisting in HepG2 cells after 24 h. To investigate the molecular basis underlying the genotoxicity attributed to CR-LAAO, we analyzed the expression profile (mRNA levels) of 44 genes involved in DNA repair and antioxidant pathways in HepG2 cells by RT2 Profiler polymerase chain reaction array. CR-LAAO altered the tumor cell expression of DNA repair genes, with two downregulated (XRCC4 and TOPBP1) and three upregulated (ERCC6, RAD52 and CDKN1) genes. In addition, two genes of the antioxidant pathway were upregulated (GPX3 and MPO), probably in an attempt to protect tumor cells from oxidative damage. In conclusion, our data suggest that CR-LAAO possesses higher binding affinity to HepG2 tumor cells than to PBMC, its genotoxic mechanism is possibly caused by the oxidative stress related to the production of H2O2, and is also capable of modulating genes related to the DNA repair system and antioxidant pathways.
Collapse
Affiliation(s)
- Tássia R Costa
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/no, CEP 14040-903, Ribeirão Preto, São Paulo, Brazil.
| | - Martin K Amstalden
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/no, CEP 14040-903, Ribeirão Preto, São Paulo, Brazil
| | - Diego L Ribeiro
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900, CEP 14049-900, Ribeirão Preto, São Paulo, Brazil
| | - Danilo L Menaldo
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/no, CEP 14040-903, Ribeirão Preto, São Paulo, Brazil
| | - Marco A Sartim
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/no, CEP 14040-903, Ribeirão Preto, São Paulo, Brazil
| | - Alexandre F Aissa
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/no, CEP 14040-903, Ribeirão Preto, São Paulo, Brazil
| | - Lusânia M G Antunes
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/no, CEP 14040-903, Ribeirão Preto, São Paulo, Brazil
| | - Suely V Sampaio
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café s/no, CEP 14040-903, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
52
|
Moga MA, Dimienescu OG, Arvătescu CA, Ifteni P, Pleş L. Anticancer Activity of Toxins from Bee and Snake Venom-An Overview on Ovarian Cancer. Molecules 2018; 23:E692. [PMID: 29562696 PMCID: PMC6017821 DOI: 10.3390/molecules23030692] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 03/11/2018] [Accepted: 03/14/2018] [Indexed: 11/16/2022] Open
Abstract
Cancer represents the disease of the millennium, a major problem in public health. The proliferation of tumor cells, angiogenesis, and the relationship between the cancer cells and the components of the extracellular matrix are important in the events of carcinogenesis, and these pathways are being used as targets for new anticancer treatments. Various venoms and their toxins have shown possible anticancer effects on human cancer cell lines, providing new perspectives in drug development. In this review, we observed the effects of natural toxins from bee and snake venom and the mechanisms through which they can inhibit the growth and proliferation of cancer cells. We also researched how several types of natural molecules from venom can sensitize ovarian cancer cells to conventional chemotherapy, with many toxins being helpful for developing new anticancer drugs. This approach could improve the efficiency of standard therapies and could allow the administration of decreased doses of chemotherapy. Natural toxins from bee and snake venom could become potential candidates for the future treatment of different types of cancer. It is important to continue these studies concerning therapeutic drugs from natural resource and, more importantly, to investigate their mechanism of action on cancer cells.
Collapse
Affiliation(s)
- Marius Alexandru Moga
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov, Brasov 500019, Romania.
| | - Oana Gabriela Dimienescu
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov, Brasov 500019, Romania.
| | - Cristian Andrei Arvătescu
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov, Brasov 500019, Romania.
| | - Petru Ifteni
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov, Brasov 500019, Romania.
| | - Liana Pleş
- Clinical Department of Obstetrics and Gynecology, The Carol Davila University of Medicine and Pharmacy, Bucharest 020021, Romania.
| |
Collapse
|
53
|
Suwannapan W, Chumnanpuen P, E-kobon T. Amplification and bioinformatics analysis of conserved FAD-binding region of L-amino acid oxidase ( LAAO) genes in gastropods compared to other organisms. Comput Struct Biotechnol J 2018; 16:98-107. [PMID: 30591829 PMCID: PMC6303269 DOI: 10.1016/j.csbj.2018.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 02/25/2018] [Accepted: 02/25/2018] [Indexed: 11/24/2022] Open
Abstract
This study aimed to investigate the conserved FAD-binding region of the L-amino acid oxidase (LAAO) genes in twelve gastropod genera commonly found in Thailand compared to those in other organisms using molecular cloning, nucleotide sequencing and bioinformatics analysis. Genomic DNA of gastropods and other invertebrates was extracted and screened using primers specific to the conserved FAD-binding region of LAAO. The amplified 143-bp fragments were cloned and sequenced. The obtained nucleotide sequences of 21 samples were aligned and phylogenetically compared to the LAAO-conserved FAD-binding regions of 210 other organisms from the NCBI database. Translated amino acid sequences of these samples were used in phylogenetics and pattern analyses. The phylogenetic trees showed clear separation of the conserved regions in fungi, invertebrates, and vertebrates. Alignment of the conserved 47-amino-acid FAD-binding region of the LAAOs showed 150 unique sequences among the 231 samples and these patterns were different from those of other flavoproteins in the amine oxidase family. An amino acid pattern analysis of five sub-regions (bFAD, FAD, FAD-GG, GG, and aGG) within the FAD-binding sequence showed high variation at the FAD-GG sub-region. Pattern analysis of secondary structures indicated the aGG sub-region as having the highest structural variation. Cluster analysis of these patterns revealed two major clusters representing the mollusc clade and the vertebrate clade. Thus, molecular phylogenetics and pattern analyses of sequence and structural variations could reflect evolutionary relatedness and possible structural conservation to maintain specific function within the FAD-binding region of the LAAOs in gastropods compared to other organisms.
Collapse
Affiliation(s)
- Wipawadee Suwannapan
- Department of Genetics, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Pramote Chumnanpuen
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Computational Biomodelling Laboratory for Agricultural Science and Technology, Kasetsart University, Bangkok 10900, Thailand
| | - Teerasak E-kobon
- Department of Genetics, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Computational Biomodelling Laboratory for Agricultural Science and Technology, Kasetsart University, Bangkok 10900, Thailand
| |
Collapse
|
54
|
Tan KK, Bay BH, Gopalakrishnakone P. L-amino acid oxidase from snake venom and its anticancer potential. Toxicon 2018; 144:7-13. [DOI: 10.1016/j.toxicon.2018.01.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 01/22/2018] [Accepted: 01/24/2018] [Indexed: 12/13/2022]
|
55
|
An Overview of l-Amino Acid Oxidase Functions from Bacteria to Mammals: Focus on the Immunoregulatory Phenylalanine Oxidase IL4I1. Molecules 2017; 22:molecules22122151. [PMID: 29206151 PMCID: PMC6149928 DOI: 10.3390/molecules22122151] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 11/23/2017] [Accepted: 11/29/2017] [Indexed: 01/04/2023] Open
Abstract
l-amino acid oxidases are flavin adenine dinucleotide-dependent enzymes present in all major kingdom of life, from bacteria to mammals. They participate in defense mechanisms by limiting the growth of most bacteria and parasites. A few mammalian LAAOs have been described, of which the enzyme “interleukin-4 induced gene 1” (IL4I1) is the best characterized. IL4I1 mainly oxidizes l-phenylalanine. It is a secreted enzyme physiologically produced by antigen presenting cells of the myeloid and B cell lineages and T helper type (Th) 17 cells. Important roles of IL4I1 in the fine control of the adaptive immune response in mice and humans have emerged during the last few years. Indeed, IL4I1 inhibits T cell proliferation and cytokine production and facilitates naïve CD4+ T-cell differentiation into regulatory T cells in vitro by limiting the capacity of T lymphocytes to respond to clonal receptor stimulation. It may also play a role in controlling the germinal center reaction for antibody production and limiting Th1 and Th17 responses. IL4I1 is expressed in tumor-associated macrophages of most human cancers and in some tumor cell types. Such expression, associated with its capacity to facilitate tumor growth by inhibiting the anti-tumor T-cell response, makes IL4I1 a new potential druggable target in the field of immunomodulation in cancer.
Collapse
|
56
|
Molla G, Melis R, Pollegioni L. Breaking the mirror: l-Amino acid deaminase, a novel stereoselective biocatalyst. Biotechnol Adv 2017; 35:657-668. [DOI: 10.1016/j.biotechadv.2017.07.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/04/2017] [Accepted: 07/30/2017] [Indexed: 12/27/2022]
|
57
|
Bhattacharjee P, Bera I, Chakraborty S, Ghoshal N, Bhattacharyya D. Aristolochic acid and its derivatives as inhibitors of snake venom L-amino acid oxidase. Toxicon 2017; 138:1-17. [PMID: 28803055 DOI: 10.1016/j.toxicon.2017.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/18/2017] [Accepted: 08/03/2017] [Indexed: 02/02/2023]
Abstract
Snake venom L-amino acid oxidase (LAAO) exerts toxicity by inducing hemorrhage, pneumorrhagia, pulmonary edema, cardiac edema, liver cell necrosis etc. Being well conserved, inhibitors of the enzyme may be synthesized using the template of the substrate, substrate binding site and features of the catalytic site of the enzyme. Previous findings showed that aristolochic acid (AA), a major constituent of Aristolochia indica, inhibits Russell's viper venom LAAO enzyme activity since, AA interacts with DNA and causes genotoxicity, derivatives of this compound were synthesized by replacing the nitro group to reduce toxicity while retaining the inhibitory potency. The interactions of AA and its derivatives with LAAO were followed by inhibition kinetics and surface plasmon resonance. Similar interactions with DNA were followed by absorption spectroscopy and atomic force microscopy. LAAO-induced cytotoxicity was evaluated by generation of reactive oxygen species (ROS), cell viability assays, confocal and epifluorescence microscopy. The hydroxyl (AA-OH) and chloro (AA-Cl) derivatives acted as inhibitors of LAAO but did not interact with DNA. The derivatives significantly reduced LAAO-induced ROS generation and cytotoxicity in human embryonic kidney (HEK 293) and hepatoma (HepG2) cell lines. Confocal images indicated that AA, AA-OH and AA-Cl interfered with the binding of LAAO to the cell membrane. AA-OH and AA-Cl significantly inhibited LAAO activity and reduced LAAO-induced cytotoxicity.
Collapse
Affiliation(s)
- Payel Bhattacharjee
- Division of Structural Biology and Bioinformatics, CSIR -Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India.
| | - Indrani Bera
- Division of Structural Biology and Bioinformatics, CSIR -Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India
| | - Subhamoy Chakraborty
- Division of Structural Biology and Bioinformatics, CSIR -Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India
| | - Nanda Ghoshal
- Division of Structural Biology and Bioinformatics, CSIR -Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India
| | - Debasish Bhattacharyya
- Division of Structural Biology and Bioinformatics, CSIR -Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India.
| |
Collapse
|
58
|
Okai M, Suwa C, Nagaoka S, Obara N, Mitsuya D, Kurihara A, Ishida M, Urano N. Neutralization of acidic drainage by Cryptococcus sp. T1 immobilized in alginate beads. Biosci Biotechnol Biochem 2017; 81:2216-2224. [PMID: 28914179 DOI: 10.1080/09168451.2017.1373586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We isolated Cryptococcus sp. T1 from Lake Tazawa's acidic water in Japan. Cryptococcus sp. T1 neutralized an acidic casamino acid solution (pH 3.0) and released ammonia from the casamino acids to aid the neutralization. The neutralization volume was estimated to be approximately 0.4 mL/h. The casamino acids' amino acids decreased (1.24→0.15 mM); ammonia increased (0.22→0.99 mM). We neutralized acidic drainage water (1 L) from a Tamagawa River neutralization plant, which was run through the column with the T1-immobilized alginate beads at a flow rate of 0.5 mL/min, and observed that the viscosity, particle size and amounts of the alginate beads affected the acidic drainage neutralization with an increase of the pH value from 5.26 to 6.61 in the last fraction. An increase in the Al concentration decreased Cryptococcus sp. T1's neutralization ability. After 48 h, the pH of acidic water with 50 mg/L Al was apparently lower than that without Al. Almost no pH increase was observed at 75 mg/L.
Collapse
Affiliation(s)
- Masahiko Okai
- a Department of Ocean Sciences , Tokyo University of Marine Science and Technology , Tokyo , Japan
| | - Chisato Suwa
- a Department of Ocean Sciences , Tokyo University of Marine Science and Technology , Tokyo , Japan
| | - Shintaro Nagaoka
- a Department of Ocean Sciences , Tokyo University of Marine Science and Technology , Tokyo , Japan
| | - Nobuo Obara
- a Department of Ocean Sciences , Tokyo University of Marine Science and Technology , Tokyo , Japan
| | - Daisuke Mitsuya
- a Department of Ocean Sciences , Tokyo University of Marine Science and Technology , Tokyo , Japan
| | - Ayako Kurihara
- b Department of Marine Biosciences , Tokyo University of Marine Science and Technology , Tokyo , Japan
| | - Masami Ishida
- a Department of Ocean Sciences , Tokyo University of Marine Science and Technology , Tokyo , Japan
| | - Naoto Urano
- a Department of Ocean Sciences , Tokyo University of Marine Science and Technology , Tokyo , Japan
| |
Collapse
|
59
|
Tan KY, Tan CH, Chanhome L, Tan NH. Comparative venom gland transcriptomics of Naja kaouthia (monocled cobra) from Malaysia and Thailand: elucidating geographical venom variation and insights into sequence novelty. PeerJ 2017; 5:e3142. [PMID: 28392982 PMCID: PMC5384570 DOI: 10.7717/peerj.3142] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 03/03/2017] [Indexed: 11/20/2022] Open
Abstract
Background The monocled cobra (Naja kaouthia) is a medically important venomous snake in Southeast Asia. Its venom has been shown to vary geographically in relation to venom composition and neurotoxic activity, indicating vast diversity of the toxin genes within the species. To investigate the polygenic trait of the venom and its locale-specific variation, we profiled and compared the venom gland transcriptomes of N. kaouthia from Malaysia (NK-M) and Thailand (NK-T) applying next-generation sequencing (NGS) technology. Methods The transcriptomes were sequenced on the Illumina HiSeq platform, assembled and followed by transcript clustering and annotations for gene expression and function. Pairwise or multiple sequence alignments were conducted on the toxin genes expressed. Substitution rates were studied for the major toxins co-expressed in NK-M and NK-T. Results and discussion The toxin transcripts showed high redundancy (41–82% of the total mRNA expression) and comprised 23 gene families expressed in NK-M and NK-T, respectively (22 gene families were co-expressed). Among the venom genes, three-finger toxins (3FTxs) predominated in the expression, with multiple sequences noted. Comparative analysis and selection study revealed that 3FTxs are genetically conserved between the geographical specimens whilst demonstrating distinct differential expression patterns, implying gene up-regulation for selected principal toxins, or alternatively, enhanced transcript degradation or lack of transcription of certain traits. One of the striking features that elucidates the inter-geographical venom variation is the up-regulation of α-neurotoxins (constitutes ∼80.0% of toxin’s fragments per kilobase of exon model per million mapped reads (FPKM)), particularly the long-chain α-elapitoxin-Nk2a (48.3%) in NK-T but only 1.7% was noted in NK-M. Instead, short neurotoxin isoforms were up-regulated in NK-M (46.4%). Another distinct transcriptional pattern observed is the exclusively and abundantly expressed cytotoxin CTX-3 in NK-T. The findings suggested correlation with the geographical variation in proteome and toxicity of the venom, and support the call for optimising antivenom production and use in the region. Besides, the current study uncovered full and partial sequences of numerous toxin genes from N. kaouthia which have not been reported hitherto; these include N. kaouthia-specific l-amino acid oxidase (LAAO), snake venom serine protease (SVSP), cystatin, acetylcholinesterase (AChE), hyaluronidase (HYA), waprin, phospholipase B (PLB), aminopeptidase (AP), neprilysin, etc. Taken together, the findings further enrich the snake toxin database and provide deeper insights into the genetic diversity of cobra venom toxins.
Collapse
Affiliation(s)
- Kae Yi Tan
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | - Choo Hock Tan
- Department of Pharmacology, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| | | | - Nget Hong Tan
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya , Kuala Lumpur , Malaysia
| |
Collapse
|
60
|
Feliciano PR, Rustiguel JK, Soares ROS, Sampaio SV, Cristina Nonato M. Crystal structure and molecular dynamics studies of L-amino acid oxidase from Bothrops atrox. Toxicon 2017; 128:50-59. [PMID: 28137621 DOI: 10.1016/j.toxicon.2017.01.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/19/2017] [Accepted: 01/23/2017] [Indexed: 11/26/2022]
Abstract
L-amino acid oxidases (LAAOs) are dimeric flavoproteins that catalyze the deamination of L-amino acid to α-keto acid, producing ammonia and hydrogen peroxide. In this study, we report the crystal structure and molecular dynamics simulations of LAAO from the venom of Bothrops atrox (BatroxLAAO). BatroxLAAO presents several biological and pharmacological properties with promising biomedical applications. BatroxLAAO structure contains the highly conserved structural pattern of LAAOs comprising a FAD-binding domain, substrate-binding domain and helical domain, and a dimeric arrangement that can be stabilized by zinc. Also, molecular dynamics results show an asymmetric behavior, and a direct communication between FAD- and substrate-binding domains of counterpart subunits. These findings shed light on the structural role of dimerization to catalytic mechanism of SV-LAAOs.
Collapse
Affiliation(s)
- Patricia R Feliciano
- Laboratório de Cristalografia de Proteínas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Joane K Rustiguel
- Laboratório de Cristalografia de Proteínas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Ricardo O S Soares
- Laboratório de Cristalografia de Proteínas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Suely V Sampaio
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - M Cristina Nonato
- Laboratório de Cristalografia de Proteínas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
61
|
Mechanism of the cytotoxic effect of l-amino acid oxidase isolated from Bothrops alternatus snake venom. Int J Biol Macromol 2016; 92:329-337. [DOI: 10.1016/j.ijbiomac.2016.07.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 07/04/2016] [Accepted: 07/05/2016] [Indexed: 01/05/2023]
|
62
|
Rodrigues EP, Soares CDP, Galvão PG, Imada EL, Simões-Araújo JL, Rouws LFM, de Oliveira ALM, Vidal MS, Baldani JI. Identification of Genes Involved in Indole-3-Acetic Acid Biosynthesis by Gluconacetobacter diazotrophicus PAL5 Strain Using Transposon Mutagenesis. Front Microbiol 2016; 7:1572. [PMID: 27774087 PMCID: PMC5053998 DOI: 10.3389/fmicb.2016.01572] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 09/20/2016] [Indexed: 02/03/2023] Open
Abstract
Gluconacetobacter diazotrophicus is a beneficial nitrogen-fixing endophyte found in association with sugarcane plants and other important crops. Beneficial effects of G. diazotrophicus on sugarcane growth and productivity have been attributed to biological nitrogen fixation process and production of phytohormones especially indole-3-acetic acid (IAA); however, information about the biosynthesis and function of IAA in G. diazotrophicus is still scarce. Therefore, the aim of this work was to identify genes and pathways involved in IAA biosynthesis in this bacterium. In our study, the screening of two independent Tn5 mutant libraries of PAL5T strain using the Salkowski colorimetric assay revealed two mutants (Gdiaa34 and Gdiaa01), which exhibited 95% less indolic compounds than the parental strain when grown in LGIP medium supplemented with L-tryptophan. HPLC chromatograms of the wild-type strain revealed the presence of IAA and of the biosynthetic intermediates indole-3-pyruvic acid (IPyA) and indole-3-lactate (ILA). In contrast, the HPLC profiles of both mutants showed no IAA but only a large peak of non-metabolized tryptophan and low levels of IPyA and ILA were detected. Molecular characterization revealed that Gdiaa01 and Gdiaa34 mutants had unique Tn5 insertions at different sites within the GDI2456 open read frame, which is predicted to encode a L-amino acid oxidase (LAAO). GDI2456 (lao gene) forms a cluster with GDI2455 and GDI2454 ORFs, which are predicted to encode a cytochrome C and an RidA protein, respectively. RT-qPCR showed that transcript levels of lao. cccA, and ridA genes were reduced in the Gdiaa01 as compared to PAL5T. In addition, rice plants inoculated with Gdiaa01 showed significantly smaller root development (length, surface area, number of forks and tips) than those plants inoculated with PAL5T. In conclusion, our study demonstrated that G. diazotrophicus PAL5T produces IAA via the IPyA pathway in cultures supplemented with tryptophan and provides evidence for the involvement of an L-amino acid oxidase gene cluster in the biosynthesis of IAA. Furthermore, we showed that the mutant strains with reduction in IAA biosynthesis ability, in consequence of the lower transcription levels of genes of the lao cluster, had remarkable effects on development of rice roots.
Collapse
Affiliation(s)
- Elisete P Rodrigues
- Laboratório de Genética de Microrganismos, Departamento de Biologia, Universidade Estadual de Londrina Londrina, Brazil
| | | | | | - Eddie L Imada
- Laboratório de Genética de Microrganismos, Departamento de Biologia, Universidade Estadual de Londrina Londrina, Brazil
| | | | | | - André L M de Oliveira
- Laboratório de Bioquímica Molecular, Departamento de Bioquímica e Biotecnologia, Universidade Estadual de Londrina Londrina, Brazil
| | | | | |
Collapse
|
63
|
Ju Y, Tong S, Gao Y, Zhao W, Liu Q, Gu Q, Xu J, Niu L, Teng M, Zhou H. Crystal structure of a membrane-bound l -amino acid deaminase from Proteus vulgaris. J Struct Biol 2016; 195:306-315. [DOI: 10.1016/j.jsb.2016.07.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 06/20/2016] [Accepted: 07/12/2016] [Indexed: 10/21/2022]
|
64
|
Leng J, Qin HL, Zhu K, Jantan I, Hussain MA, Sher M, Amjad MW, Naeem-ul-Hassan M, Ahmad W, Bukhari SNA. Evaluation of multifunctional synthetic tetralone derivatives for treatment of Alzheimer's disease. Chem Biol Drug Des 2016; 88:889-898. [DOI: 10.1111/cbdd.12822] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 07/02/2016] [Accepted: 07/12/2016] [Indexed: 01/30/2023]
Affiliation(s)
- Jing Leng
- Department of Pharmaceutical Engineering; School of Chemistry; Chemical Engineering and Life Science; Wuhan University of Technology; Wuhan China
| | - Hua-Li Qin
- Department of Pharmaceutical Engineering; School of Chemistry; Chemical Engineering and Life Science; Wuhan University of Technology; Wuhan China
| | - Kaicheng Zhu
- Department of Pharmaceutical Engineering; School of Chemistry; Chemical Engineering and Life Science; Wuhan University of Technology; Wuhan China
| | - Ibrahim Jantan
- Drug and Herbal Research Centre; Faculty of Pharmacy; Universiti Kebangsaan Malaysia; Kuala Lumpur Malaysia
| | | | - Muhammad Sher
- Department of Chemistry; University of Sargodha; Sargodha Pakistan
| | - Muhammad Wahab Amjad
- Drug and Herbal Research Centre; Faculty of Pharmacy; Universiti Kebangsaan Malaysia; Kuala Lumpur Malaysia
| | | | - Waqas Ahmad
- Drug and Herbal Research Centre; Faculty of Pharmacy; Universiti Kebangsaan Malaysia; Kuala Lumpur Malaysia
| | - Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Engineering; School of Chemistry; Chemical Engineering and Life Science; Wuhan University of Technology; Wuhan China
- Drug and Herbal Research Centre; Faculty of Pharmacy; Universiti Kebangsaan Malaysia; Kuala Lumpur Malaysia
| |
Collapse
|
65
|
Füller JJ, Röpke R, Krausze J, Rennhack KE, Daniel NP, Blankenfeldt W, Schulz S, Jahn D, Moser J. Biosynthesis of Violacein, Structure and Function of l-Tryptophan Oxidase VioA from Chromobacterium violaceum. J Biol Chem 2016; 291:20068-84. [PMID: 27466367 DOI: 10.1074/jbc.m116.741561] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Indexed: 11/06/2022] Open
Abstract
Violacein is a natural purple pigment of Chromobacterium violaceum with potential medical applications as antimicrobial, antiviral, and anticancer drugs. The initial step of violacein biosynthesis is the oxidative conversion of l-tryptophan into the corresponding α-imine catalyzed by the flavoenzyme l-tryptophan oxidase (VioA). A substrate-related (3-(1H-indol-3-yl)-2-methylpropanoic acid) and a product-related (2-(1H-indol-3-ylmethyl)prop-2-enoic acid) competitive VioA inhibitor was synthesized for subsequent kinetic and x-ray crystallographic investigations. Structures of the binary VioA·FADH2 and of the ternary VioA·FADH2·2-(1H-indol-3-ylmethyl)prop-2-enoic acid complex were resolved. VioA forms a "loosely associated" homodimer as indicated by small-angle x-ray scattering experiments. VioA belongs to the glutathione reductase family 2 of FAD-dependent oxidoreductases according to the structurally conserved cofactor binding domain. The substrate-binding domain of VioA is mainly responsible for the specific recognition of l-tryptophan. Other canonical amino acids were efficiently discriminated with a minor conversion of l-phenylalanine. Furthermore, 7-aza-tryptophan, 1-methyl-tryptophan, 5-methyl-tryptophan, and 5-fluoro-tryptophan were efficient substrates of VioA. The ternary product-related VioA structure indicated involvement of protein domain movement during enzyme catalysis. Extensive structure-based mutagenesis in combination with enzyme kinetics (using l-tryptophan and substrate analogs) identified Arg(64), Lys(269), and Tyr(309) as key catalytic residues of VioA. An increased enzyme activity of protein variant H163A in the presence of l-phenylalanine indicated a functional role of His(163) in substrate binding. The combined structural and mutational analyses lead to the detailed understanding of VioA substrate recognition. Related strategies for the in vivo synthesis of novel violacein derivatives are discussed.
Collapse
Affiliation(s)
| | - René Röpke
- the Institute of Organic Chemistry, Technische Universität Braunschweig, Hagenring 30, D-38106 Braunschweig, and
| | - Joern Krausze
- the Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, D-38124 Braunschweig, Germany
| | | | | | - Wulf Blankenfeldt
- the Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, D-38124 Braunschweig, Germany Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Spielmannstrasse 7, D-38106 Braunschweig
| | - Stefan Schulz
- the Institute of Organic Chemistry, Technische Universität Braunschweig, Hagenring 30, D-38106 Braunschweig, and
| | | | | |
Collapse
|
66
|
Melani RD, Skinner OS, Fornelli L, Domont GB, Compton PD, Kelleher NL. Mapping Proteoforms and Protein Complexes From King Cobra Venom Using Both Denaturing and Native Top-down Proteomics. Mol Cell Proteomics 2016; 15:2423-34. [PMID: 27178327 DOI: 10.1074/mcp.m115.056523] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Indexed: 11/06/2022] Open
Abstract
Characterizing whole proteins by top-down proteomics avoids a step of inference encountered in the dominant bottom-up methodology when peptides are assembled computationally into proteins for identification. The direct interrogation of whole proteins and protein complexes from the venom of Ophiophagus hannah (king cobra) provides a sharply clarified view of toxin sequence variation, transit peptide cleavage sites and post-translational modifications (PTMs) likely critical for venom lethality. A tube-gel format for electrophoresis (called GELFrEE) and solution isoelectric focusing were used for protein fractionation prior to LC-MS/MS analysis resulting in 131 protein identifications (18 more than bottom-up) and a total of 184 proteoforms characterized from 14 protein toxin families. Operating both GELFrEE and mass spectrometry to preserve non-covalent interactions generated detailed information about two of the largest venom glycoprotein complexes: the homodimeric l-amino acid oxidase (∼130 kDa) and the multichain toxin cobra venom factor (∼147 kDa). The l-amino acid oxidase complex exhibited two clusters of multiproteoform complexes corresponding to the presence of 5 or 6 N-glycans moieties, each consistent with a distribution of N-acetyl hexosamines. Employing top-down proteomics in both native and denaturing modes provides unprecedented characterization of venom proteoforms and their complexes. A precise molecular inventory of venom proteins will propel the study of snake toxin variation and the targeted development of new antivenoms or other biotherapeutics.
Collapse
Affiliation(s)
- Rafael D Melani
- From the ‡Proteomics Unit, Rio de Janeiro Proteomics Network, Departamento de Bioquímica. Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-909, Brazil; §Departments of Chemistry and Molecular Biosciences, the Chemistry of Life Processes Institute, and the Proteomics Center of Excellence, Northwestern University, Evanston, Illinois, 60208
| | - Owen S Skinner
- §Departments of Chemistry and Molecular Biosciences, the Chemistry of Life Processes Institute, and the Proteomics Center of Excellence, Northwestern University, Evanston, Illinois, 60208
| | - Luca Fornelli
- §Departments of Chemistry and Molecular Biosciences, the Chemistry of Life Processes Institute, and the Proteomics Center of Excellence, Northwestern University, Evanston, Illinois, 60208
| | - Gilberto B Domont
- From the ‡Proteomics Unit, Rio de Janeiro Proteomics Network, Departamento de Bioquímica. Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-909, Brazil;
| | - Philip D Compton
- §Departments of Chemistry and Molecular Biosciences, the Chemistry of Life Processes Institute, and the Proteomics Center of Excellence, Northwestern University, Evanston, Illinois, 60208
| | - Neil L Kelleher
- §Departments of Chemistry and Molecular Biosciences, the Chemistry of Life Processes Institute, and the Proteomics Center of Excellence, Northwestern University, Evanston, Illinois, 60208
| |
Collapse
|
67
|
Motta P, Molla G, Pollegioni L, Nardini M. Structure-Function Relationships in l-Amino Acid Deaminase, a Flavoprotein Belonging to a Novel Class of Biotechnologically Relevant Enzymes. J Biol Chem 2016; 291:10457-75. [PMID: 27022028 DOI: 10.1074/jbc.m115.703819] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Indexed: 01/11/2023] Open
Abstract
l-Amino acid deaminase from Proteus myxofaciens (PmaLAAD) is a membrane flavoenzyme that catalyzes the deamination of neutral and aromatic l-amino acids into α-keto acids and ammonia. PmaLAAD does not use dioxygen to re-oxidize reduced FADH2 and thus does not produce hydrogen peroxide; instead, it uses a cytochrome b-like protein as an electron acceptor. Although the overall fold of this enzyme resembles that of known amine or amino acid oxidases, it shows the following specific structural features: an additional novel α+β subdomain placed close to the putative transmembrane α-helix and to the active-site entrance; an FAD isoalloxazine ring exposed to solvent; and a large and accessible active site suitable to bind large hydrophobic substrates. In addition, PmaLAAD requires substrate-induced conformational changes of part of the active site, particularly in Arg-316 and Phe-318, to achieve the correct geometry for catalysis. These studies are expected to pave the way for rationally improving the versatility of this flavoenzyme, which is critical for biocatalysis of enantiomerically pure amino acids.
Collapse
Affiliation(s)
- Paolo Motta
- From the Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi deII'Insubria, via J. H. Dunant 3, 21100 Varese
| | - Gianluca Molla
- From the Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi deII'Insubria, via J. H. Dunant 3, 21100 Varese, The Protein Factory, Politecnico di Milano and Università degli Studi deII'Insubria, 21100 Varese, and
| | - Loredano Pollegioni
- From the Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi deII'Insubria, via J. H. Dunant 3, 21100 Varese, The Protein Factory, Politecnico di Milano and Università degli Studi deII'Insubria, 21100 Varese, and
| | - Marco Nardini
- the Dipartimento di Bioscienze, Università degli Studi di Milano, via Celoria 26, 20133 Milano, Italy
| |
Collapse
|
68
|
Fitzpatrick PF, Chadegani F, Zhang S, Roberts KM, Hinck CS. Mechanism of the Flavoprotein L-Hydroxynicotine Oxidase: Kinetic Mechanism, Substrate Specificity, Reaction Product, and Roles of Active-Site Residues. Biochemistry 2016; 55:697-703. [PMID: 26744768 PMCID: PMC4738163 DOI: 10.1021/acs.biochem.5b01325] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The flavoprotein L-hydroxynicotine oxidase (LHNO) catalyzes an early step in the bacterial catabolism of nicotine. Although the structure of the enzyme establishes that it is a member of the monoamine oxidase family, LHNO is generally accepted to oxidize a carbon-carbon bond in the pyrrolidine ring of the substrate and has been proposed to catalyze the subsequent tautomerization and hydrolysis of the initial oxidation product to yield 6-hydroxypseudooxynicotine [Kachalova, G., et al. (2011) Proc. Natl. Acad. Sci. U.S.A. 108, 4800-4805]. Analysis of the product of the enzyme from Arthrobacter nicotinovorans by nuclear magnetic resonance and continuous-flow mass spectrometry establishes that the enzyme catalyzes the oxidation of the pyrrolidine carbon-nitrogen bond, the expected reaction for a monoamine oxidase, and that hydrolysis of the amine to form 6-hydroxypseudooxynicotine is nonenzymatic. On the basis of the kcat/Km and kred values for (S)-hydroxynicotine and several analogues, the methyl group contributes only marginally (∼ 0.5 kcal/mol) to transition-state stabilization, while the hydroxyl oxygen and pyridyl nitrogen each contribute ∼ 4 kcal/mol. The small effects on activity of mutagenesis of His187, Glu300, or Tyr407 rule out catalytic roles for all three of these active-site residues.
Collapse
Affiliation(s)
- Paul F. Fitzpatrick
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229
| | - Fatemeh Chadegani
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229
| | - Shengnan Zhang
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229
| | - Kenneth M. Roberts
- Department of Chemistry & Physics, University of South Carolina Aiken, Aiken, SC 29801
| | - Cynthia S. Hinck
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229
| |
Collapse
|
69
|
Murray PJ. Amino acid auxotrophy as a system of immunological control nodes. Nat Immunol 2016; 17:132-9. [PMID: 26784254 PMCID: PMC4893777 DOI: 10.1038/ni.3323] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 10/13/2015] [Indexed: 02/07/2023]
Abstract
Cells of the immune system are auxotrophs for most amino acids, including several nonessential ones. Arginine and tryptophan are used within the regulatory immune networks to control proliferation and function through pathways that actively deplete the amino acid from the microenvironment or that create regulatory molecules such as nitric oxide or kynurenines. How immune cells integrate information about essential amino acid supplies and then transfer these signals to growth and activation pathways remains unclear but has potential for pathway discovery about amino sensing. In applied research, strategies to harness amino acid auxotrophy so as to block cancerous lymphocyte growth have been attempted for decades with limited success. Emerging insights about amino acid metabolism may lead to new strategies in clinical medicine whereby both amino acid auxotrophy and the immunoregulatory pathways controlled by amino acids can be manipulated.
Collapse
Affiliation(s)
- Peter J Murray
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
70
|
Burin SM, Ghisla S, Ouchida AT, Aissa AF, Coelho MGB, Costa TR, Marsola APZC, Pinto-Simões B, Antunes LMG, Curti C, Sampaio SV, de Castro FA. CR-LAAO antileukemic effect against Bcr-Abl(+) cells is mediated by apoptosis and hydrogen peroxide. Int J Biol Macromol 2016; 86:309-20. [PMID: 26812110 DOI: 10.1016/j.ijbiomac.2016.01.069] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 01/15/2016] [Accepted: 01/20/2016] [Indexed: 02/01/2023]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm characterized by the presence of the Bcr-Abl tyrosine kinase protein, which confers resistance to apoptosis in leukemic cells. Tyrosine kinase inhibitors (TKIs) are effectively used to treat CML; however, CML patients in the advanced (CML-AP) and chronic (CML-CP) phases of the disease are usually resistant to TKI therapy. Thus, it is necessary to seek for novel agents to treat CML, such as the enzyme l-amino acid oxidase from Calloselasma rhodostoma (CR-LAAO) snake venom. We examined the antitumor effect of CR-LAAO in Bcr-Abl(+) cell lines and peripheral blood mononuclear cells (PBMC) from healthy subjects and CML patients. CR-LAAO was more cytotoxic towards Bcr-Abl(+) cell lines than towards healthy subjects' PBMC. The H2O2 produced during the enzymatic action of CR-LAAO mediated its cytotoxic effect. The CR-LAAO induced apoptosis in Bcr-Abl(+) cells, as detected by caspases 3, 8, and 9 activation, loss of mitochondrial membrane potential, and DNA damage. CR-LAAO elicited apoptosis in PBMC from CML-CP patients without TKI treatment more strongly than in PBMC from healthy subjects and TKI-treated CML-CP and CML-AP patients. The antitumor effect of CR-LAAO against Bcr-Abl(+) cells makes this toxin a promising candidate to CML therapy.
Collapse
Affiliation(s)
- Sandra Mara Burin
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, SP, Brazil.
| | - Sandro Ghisla
- Department of Biology, University of Konstanz, Konstanz, Germany.
| | - Amanda Tomie Ouchida
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo-USP, Ribeirão Preto, SP, Brazil.
| | - Alexandre Ferro Aissa
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, SP, Brazil.
| | - Maria Gabriela Berzoti Coelho
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, SP, Brazil.
| | - Tássia Rafaella Costa
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, SP, Brazil.
| | - Ana Paula Zambuzi Cardoso Marsola
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, SP, Brazil.
| | - Belinda Pinto-Simões
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo-USP, Ribeirão Preto, SP, Brazil.
| | - Lusânia Maria Greggi Antunes
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, SP, Brazil.
| | - Carlos Curti
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, SP, Brazil.
| | - Suely Vilela Sampaio
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, SP, Brazil.
| | - Fabíola Attié de Castro
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
71
|
Abstract
Cells of the immune system are auxotrophs for most amino acids, including several nonessential ones. Arginine and tryptophan are used within the regulatory immune networks to control proliferation and function through pathways that actively deplete the amino acid from the microenvironment or that create regulatory molecules such as nitric oxide or kynurenines. How immune cells integrate information about essential amino acid supplies and then transfer these signals to growth and activation pathways remains unclear but has potential for pathway discovery about amino sensing. In applied research, strategies to harness amino acid auxotrophy so as to block cancerous lymphocyte growth have been attempted for decades with limited success. Emerging insights about amino acid metabolism may lead to new strategies in clinical medicine whereby both amino acid auxotrophy and the immunoregulatory pathways controlled by amino acids can be manipulated.
Collapse
|
72
|
Alterations of the immunosuppressive IL4I1 enzyme activity induced by naturally occurring SNP/mutations. Genes Immun 2015; 17:148-52. [DOI: 10.1038/gene.2015.55] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 10/26/2015] [Accepted: 11/11/2015] [Indexed: 11/08/2022]
|
73
|
El Hakim A, Salama W, Hamed M, Ali A, Ibrahim N. Heterodimeric l-amino acid oxidase enzymes from Egyptian Cerastes cerastes venom: Purification, biochemical characterization and partial amino acid sequencing. J Genet Eng Biotechnol 2015; 13:165-176. [PMID: 30647580 PMCID: PMC6299811 DOI: 10.1016/j.jgeb.2015.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 09/02/2015] [Accepted: 09/06/2015] [Indexed: 11/24/2022]
Abstract
Two l-amino acid oxidase enzyme isoforms, Cc-LAAOI and Cc-LAAOII were purified to apparent homogeneity from Cerastes cerastes venom in a sequential two-step chromatographic protocol including; gel filtration and anion exchange chromatography. The native molecular weights of the isoforms were 115 kDa as determined by gel filtration on calibrated Sephacryl S-200 column, while the monomeric molecular weights of the enzymes were, 60, 56 kDa and 60, 53 kDa for LAAOI and LAAOII, respectively. The tryptic peptides of the two isoforms share high sequence homology with other snake venom l-amino acid oxidases. The optimal pH and temperature values of Cc-LAAOI and Cc-LAAOII were 7.8, 50 °C and 7, 60 °C, respectively. The two isoenzymes were thermally stable up to 70 °C. The K m and V max values were 0.67 mM, 0.135 μmol/min for LAAOI and 0.82 mM, 0.087 μmol/min for LAAOII. Both isoenzymes displayed high catalytic preference to long-chain, hydrophobic and aromatic amino acids. The Mn2 + ion markedly increased the LAAO activity for both purified isoforms, while Na+, K+, Ca2 +, Mg2 + and Ba2 + ions showed a non-significant increase in the enzymatic activity of both isoforms. Furthermore, Zn2 +, Ni2 +, Co2 +, Cu2 + and AL3 + ions markedly inhibited the LAAOI and LAAOII activities. l-Cysteine and reduced glutathione completely inhibited the LAAO activity of both isoenzymes, whereas, β-mercaptoethanol, O-phenanthroline and PMSF completely inhibited the enzymatic activity of LAAOII. Furthermore, iodoacitic acid inhibited the enzymatic activity of LAAOII by 46% and had no effect on the LAAOI activity.
Collapse
Affiliation(s)
- A.E. El Hakim
- Molecular Biology Department, National Research Centre, 33 Bohouth St. (former El Tahrir St.), Dokki, Giza, Egypt1
| | - W.H. Salama
- Molecular Biology Department, National Research Centre, 33 Bohouth St. (former El Tahrir St.), Dokki, Giza, Egypt1
| | - M.B. Hamed
- Molecular Biology Department, National Research Centre, 33 Bohouth St. (former El Tahrir St.), Dokki, Giza, Egypt1
| | - A.A. Ali
- Molecular Biology Department, National Research Centre, 33 Bohouth St. (former El Tahrir St.), Dokki, Giza, Egypt1
- Durham University, School of Biological and Biomedical Sciences, Durham DH1 3LE, United Kingdom
- Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom
| | - N.M. Ibrahim
- Molecular Biology Department, National Research Centre, 33 Bohouth St. (former El Tahrir St.), Dokki, Giza, Egypt1
| |
Collapse
|
74
|
Tan CH, Tan KY, Fung SY, Tan NH. Venom-gland transcriptome and venom proteome of the Malaysian king cobra (Ophiophagus hannah). BMC Genomics 2015; 16:687. [PMID: 26358635 PMCID: PMC4566206 DOI: 10.1186/s12864-015-1828-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 08/07/2015] [Indexed: 02/01/2023] Open
Abstract
Background The king cobra (Ophiophagus hannah) is widely distributed throughout many parts of Asia. This study aims to investigate the complexity of Malaysian Ophiophagus hannah (MOh) venom for a better understanding of king cobra venom variation and its envenoming pathophysiology. The venom gland transcriptome was investigated using the Illumina HiSeq™ platform, while the venom proteome was profiled by 1D-SDS-PAGE-nano-ESI-LCMS/MS. Results Transcriptomic results reveal high redundancy of toxin transcripts (3357.36 FPKM/transcript) despite small cluster numbers, implying gene duplication and diversification within restricted protein families. Among the 23 toxin families identified, three-finger toxins (3FTxs) and snake-venom metalloproteases (SVMPs) have the most diverse isoforms. These 2 toxin families are also the most abundantly transcribed, followed in descending order by phospholipases A2 (PLA2s), cysteine-rich secretory proteins (CRISPs), Kunitz-type inhibitors (KUNs), and L-amino acid oxidases (LAAOs). Seventeen toxin families exhibited low mRNA expression, including hyaluronidase, DPP-IV and 5’-nucleotidase that were not previously reported in the venom-gland transcriptome of a Balinese O. hannah. On the other hand, the MOh proteome includes 3FTxs, the most abundantly expressed proteins in the venom (43 % toxin sbundance). Within this toxin family, there are 6 long-chain, 5 short-chain and 2 non-conventional 3FTx. Neurotoxins comprise the major 3FTxs in the MOh venom, consistent with rapid neuromuscular paralysis reported in systemic envenoming. The presence of toxic enzymes such as LAAOs, SVMPs and PLA2 would explain tissue inflammation and necrotising destruction in local envenoming. Dissimilarities in the subtypes and sequences between the neurotoxins of MOh and Naja kaouthia (monocled cobra) are in agreement with the poor cross-neutralization activity of N. kaouthia antivenom used against MOh venom. Besides, the presence of cobra venom factor, nerve growth factors, phosphodiesterase, 5’-nucleotidase, and DPP-IV in the venom proteome suggests its probable hypotensive action in subduing prey. Conclusion This study reports the diversity and abundance of toxins in the venom of the Malaysian king cobra (MOh). The results correlate with the pathophysiological actions of MOh venom, and dispute the use of Naja cobra antivenoms to treat MOh envenomation. The findings also provide a deeper insight into venom variations due to geography, which is crucial for the development of a useful pan-regional antivenom. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1828-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Choo Hock Tan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| | - Kae Yi Tan
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| | - Shin Yee Fung
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| | - Nget Hong Tan
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| |
Collapse
|
75
|
Evaluating the microbicidal, antiparasitic and antitumor effects of CR-LAAO from Calloselasma rhodostoma venom. Int J Biol Macromol 2015; 80:489-97. [DOI: 10.1016/j.ijbiomac.2015.07.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 07/02/2015] [Accepted: 07/03/2015] [Indexed: 01/25/2023]
|
76
|
Hoag MR, Roman J, Beaupre BA, Silvaggi NR, Moran GR. Bacterial Renalase: Structure and Kinetics of an Enzyme with 2- and 6-Dihydro-β-NAD(P) Oxidase Activity from Pseudomonas phaseolicola. Biochemistry 2015; 54:3791-802. [PMID: 26016690 DOI: 10.1021/acs.biochem.5b00451] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Despite a lack of convincing in vitro evidence and a number of sound refutations, it is widely accepted that renalase is an enzyme unique to animals that catalyzes the oxidative degradation of catecholamines in blood in order to lower vascular tone. Very recently, we identified isomers of β-NAD(P)H as substrates for renalase (Beaupre, B. A. et al. (2015) Biochemistry, 54, 795-806). These molecules carry the hydride equivalent on the 2 or 6 position of the nicotinamide base and presumably arise in nonspecific redox reactions of nicotinamide dinucleotides. Renalase serves to rapidly oxidize these isomers to form β-NAD(P)⁺ and then pass the electrons to dioxygen, forming H₂O₂. We have also shown that these substrate molecules are highly inhibitory to dehydrogenase enzymes and thus have proposed an intracellular metabolic role for this enzyme. Here, we identify a renalase from an organism without a circulatory system. This bacterial form of renalase has the same substrate specificity profile as that of human renalase but, in terms of binding constant (K(d)), shows a marked preference for substrates derived from β-NAD⁺. 2-dihydroNAD(P) substrates reduce the enzyme with rate constants (k(red)) that greatly exceed those for 6-dihydroNAD(P) substrates. Taken together, k(red)/K(d) values indicate a minimum 20-fold preference for 2DHNAD. We also offer the first structures of a renalase in complex with catalytically relevant ligands β-NAD⁺ and β-NADH (the latter being an analogue of the substrate(s)). These structures show potential electrostatic repulsion interactions with the product and a unique binding orientation for the substrate nicotinamide base that is consistent with the identified activity.
Collapse
Affiliation(s)
- Matthew R Hoag
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 North Cramer Street, Milwaukee, Wisconsin 53211-3209, United States
| | - Joseph Roman
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 North Cramer Street, Milwaukee, Wisconsin 53211-3209, United States
| | - Brett A Beaupre
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 North Cramer Street, Milwaukee, Wisconsin 53211-3209, United States
| | - Nicholas R Silvaggi
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 North Cramer Street, Milwaukee, Wisconsin 53211-3209, United States
| | - Graham R Moran
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 North Cramer Street, Milwaukee, Wisconsin 53211-3209, United States
| |
Collapse
|
77
|
Kitani Y, Fernandes JMO, Kiron V. Identification of the Atlantic cod L-amino acid oxidase and its alterations following bacterial exposure. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 50:116-120. [PMID: 25681742 DOI: 10.1016/j.dci.2015.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 02/08/2015] [Accepted: 02/08/2015] [Indexed: 06/04/2023]
Abstract
Antibacterial factors that are present in epidermal mucus of fish have a potential role in the first line of host defence to bacterial pathogens. This study reports the identification of L-amino acid oxidase (LAO) in Atlantic cod (GmLao) and the changes in the molecule following bacterial exposure. The gmlao transcripts and LAO activity were present on both the body surface and in the internal organs of the fish. Relative mRNA level of gmlao increased significantly in the gills, the spleen and the head kidney (up to 8-fold) of fish that were challenged with the pathogen Vibrio anguillarum. The gmlao expression in skin was 4-fold higher in challenged fish. Our data indicate that LAO may be an important effector of antibacterial defence in Atlantic cod.
Collapse
Affiliation(s)
- Yoichiro Kitani
- Faculty of Biosciences and Aquaculture, University of Nordland, 8049 Bodø, Norway.
| | - Jorge M O Fernandes
- Faculty of Biosciences and Aquaculture, University of Nordland, 8049 Bodø, Norway
| | - Viswanath Kiron
- Faculty of Biosciences and Aquaculture, University of Nordland, 8049 Bodø, Norway
| |
Collapse
|
78
|
The catalytic function of renalase: A decade of phantoms. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1864:177-86. [PMID: 25900362 DOI: 10.1016/j.bbapap.2015.04.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/08/2015] [Accepted: 04/10/2015] [Indexed: 12/31/2022]
Abstract
Ten years after the initial identification of human renalase the first genuinely catalytic substrates have been identified. Throughout the prior decade a consensus belief that renalase is produced predominantly by the kidney and catalytically oxidizes catecholamines in order to lower blood pressure and slow the heart has prevailed. This belief was, however, based on fundamentally flawed scientific observations that did not include control reactions to account for the well-known autoxidation of catecholamines in oxygenated solutions. Nonetheless, the initial claims have served as the kernel for a rapidly expanding body of research largely predicated on the belief that catecholamines are substrates for this enzyme. The proliferation of scientific studies pertaining to renalase as a hormone has proceeded unabated despite well-reasoned expressions of dissent that have indicated the deficiencies of the initial observations and other inconsistencies. Our group has very recently identified isomeric forms of β-NAD(P)H as substrates for renalase. These substrates arise from non-specific reduction of β-NAD(P)(+) that forms β-4-dihydroNAD(P) (β-NAD(P)H), β-2-dihydroNAD(P) and β-6-dihydroNAD(P); the latter two being substrates for renalase. Renalase oxidizes these substrates with rate constants that are up to 10(4)-fold faster than any claimed for catecholamines. The electrons harvested are delivered to dioxygen via the enzyme's FAD cofactor forming both H2O2 and β-NAD(P)(+) as products. It would appear that the metabolic purpose of this chemistry is to alleviate the inhibitory effect of β-2-dihydroNAD(P) and β-6-dihydroNAD(P) on primary metabolism dehydrogenase enzymes. The identification of this genuinely catalytic activity for renalase calls for re-evaluation of much of the research of this enzyme, in which definitive links between renalase catecholamine consumption and physiological responses were reported. This article is part of a Special Issue entitled: Physiological enzymology and protein functions.
Collapse
|
79
|
Amano M, Mizuguchi H, Sano T, Kondo H, Shinyashiki K, Inagaki J, Tamura T, Kawaguchi T, Kusakabe H, Imada K, Inagaki K. Recombinant expression, molecular characterization and crystal structure of antitumor enzyme, L-lysine α-oxidase from Trichoderma viride. J Biochem 2015; 157:549-59. [PMID: 25648943 DOI: 10.1093/jb/mvv012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 01/15/2015] [Indexed: 11/13/2022] Open
Abstract
L-Lysine α-oxidase (LysOX) from Trichoderma viride is a homodimeric 112 kDa flavoenzyme that catalyzes the oxidative deamination of L-lysine to form α-keto-ε-aminocaproate. LysOX severely inhibited growth of cancer cells but showed relatively low cytotoxicity for normal cells. We have determined the cDNA nucleotide sequence encoding LysOX from T. viride. The full-length cDNA consists of 2,119 bp and encodes a possible signal peptide (Met1-Arg77) and the mature protein (Ala78-Ile617). The LysOX gene have been cloned and heterologously expressed in Streptomyces lividans TK24 with the enzyme activity up to 9.8 U/ml. The enzymatic properties of the purified recombinant LysOX, such as substrate specificity and thermal stability, are same as those of native LysOX. The crystal structure of LysOX at 1.9 Å resolution revealed that the overall structure is similar to that of snake venom L-amino acid oxidase (LAAO), and the residues involved in the interaction with the amino or carboxy group of the substrate are structurally conserved. However, the entrance and the inner surface structures of the funnel to the active site, as well as the residues involved in the substrate side-chain recognition, are distinct from LAAOs. These structural differences well explain the unique substrate specificity of LysOX.
Collapse
Affiliation(s)
- Marie Amano
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan; Department of Macromolecular Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; and Enzyme Sensor Co., Ltd., Tsukuba, Ibaraki 305-0047, Japan
| | - Haruka Mizuguchi
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan; Department of Macromolecular Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; and Enzyme Sensor Co., Ltd., Tsukuba, Ibaraki 305-0047, Japan
| | - Tadahisa Sano
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan; Department of Macromolecular Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; and Enzyme Sensor Co., Ltd., Tsukuba, Ibaraki 305-0047, Japan
| | - Hiroki Kondo
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan; Department of Macromolecular Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; and Enzyme Sensor Co., Ltd., Tsukuba, Ibaraki 305-0047, Japan
| | - Kengo Shinyashiki
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan; Department of Macromolecular Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; and Enzyme Sensor Co., Ltd., Tsukuba, Ibaraki 305-0047, Japan
| | - Junko Inagaki
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan; Department of Macromolecular Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; and Enzyme Sensor Co., Ltd., Tsukuba, Ibaraki 305-0047, Japan
| | - Takashi Tamura
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan; Department of Macromolecular Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; and Enzyme Sensor Co., Ltd., Tsukuba, Ibaraki 305-0047, Japan
| | - Tatsuya Kawaguchi
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan; Department of Macromolecular Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; and Enzyme Sensor Co., Ltd., Tsukuba, Ibaraki 305-0047, Japan
| | - Hitoshi Kusakabe
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan; Department of Macromolecular Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; and Enzyme Sensor Co., Ltd., Tsukuba, Ibaraki 305-0047, Japan
| | - Katsumi Imada
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan; Department of Macromolecular Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; and Enzyme Sensor Co., Ltd., Tsukuba, Ibaraki 305-0047, Japan
| | - Kenji Inagaki
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan; Department of Macromolecular Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; and Enzyme Sensor Co., Ltd., Tsukuba, Ibaraki 305-0047, Japan
| |
Collapse
|
80
|
Molla G, Nardini M, Motta P, D'Arrigo P, Panzeri W, Pollegioni L. Aminoacetone oxidase from Streptococcus oligofermentans belongs to a new three-domain family of bacterial flavoproteins. Biochem J 2014; 464:387-99. [PMID: 25269103 DOI: 10.1042/bj20140972] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
The aaoSo gene from Streptococcus oligofermentans encodes a 43 kDa flavoprotein, aminoacetone oxidase (SoAAO), which was reported to possess a low catalytic activity against several different L-amino acids; accordingly, it was classified as an L-amino acid oxidase. Subsequently, SoAAO was demonstrated to oxidize aminoacetone (a pro-oxidant metabolite), with an activity ~25-fold higher than the activity displayed on L-lysine, thus lending support to the assumption of aminoacetone as the preferred substrate. In the present study, we have characterized the SoAAO structure-function relationship. SoAAO is an FAD-containing enzyme that does not possess the classical properties of the oxidase/dehydrogenase class of flavoproteins (i.e. no flavin semiquinone formation is observed during anaerobic photoreduction as well as no reaction with sulfite) and does not show a true L-amino acid oxidase activity. From a structural point of view, SoAAO belongs to a novel protein family composed of three domains: an α/β domain corresponding to the FAD-binding domain, a β-domain partially modulating accessibility to the coenzyme, and an additional α-domain. Analysis of the reaction products of SoAAO on aminoacetone showed 2,5-dimethylpyrazine as the main product; we propose that condensation of two aminoacetone molecules yields 3,6-dimethyl-2,5-dihydropyrazine that is subsequently oxidized to 2,5-dimethylpyrazine. The ability of SoAAO to bind two molecules of the substrate analogue O-methylglycine ligand is thought to facilitate the condensation reaction. A specialized role for SoAAO in the microbial defence mechanism related to aminoacetone catabolism through a pathway yielding dimethylpyrazine derivatives instead of methylglyoxal can be proposed.
Collapse
Affiliation(s)
- Gianluca Molla
- *Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi deII'Insubria, via J.H. Dunant 3, 21100 Varese, ltaly
| | - Marco Nardini
- ‡Dipartimento di Bioscienze, Università degli Studi di Milano, 20133 Milano, Italy
| | - Paolo Motta
- *Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi deII'Insubria, via J.H. Dunant 3, 21100 Varese, ltaly
| | - Paola D'Arrigo
- †The Protein Factory, Centro Interuniversitario di Biotecnologie Proteiche, Politecnico di Milano, ICRM CNR Milano, and Università degli Studi deII'Insubria, Varese, Italy
| | - Walter Panzeri
- ║CNR-Istituto di Chimica del Riconoscimento Molecolare, Politecnico di Milano, via Mancinelli 7, 20131 Milano, Italy
| | - Loredano Pollegioni
- *Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi deII'Insubria, via J.H. Dunant 3, 21100 Varese, ltaly
| |
Collapse
|
81
|
Tani Y, Omatsu K, Saito S, Miyake R, Kawabata H, Ueda M, Mihara H. Heterologous expression of l-lysine α-oxidase from Scomber japonicus in Pichia pastoris and functional characterization of the recombinant enzyme. J Biochem 2014; 157:201-10. [PMID: 25359785 DOI: 10.1093/jb/mvu064] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Fish have a complex self-defense mechanism against microbial invasion. Recently, l-lysine α-oxidases have been identified from a number of fish species as a novel type of antibacterial protein in the integument. These enzymes exhibit strict substrate specificity for l-lysine, but the underlying mechanisms and details of their catalytic properties remain unknown. In this study, a synthetic gene coding for Scomber japonicus l-lysine α-oxidase, originally termed AIP (for apoptosis-inducing protein), was expressed in Pichia pastoris, and the recombinant enzyme (rAIP) was purified and characterized. rAIP exhibited essentially the same substrate specificity as the native enzyme, catalyzing the oxidative deamination of l-lysine as an exclusive substrate. rAIP was N-glycosylated and remained active over a wide range of pH, with an optimal pH of 7.5. The enzyme was stable in the pH range from 4.5 to 10.0 and was thermally stable up to 60°C. A molecular modelling of rAIP and a comparative structure/sequence analysis with homologous enzymes indicate that Asp(220) and Asp(320) are the substrate-binding residues that are likely to confer exclusive substrate specificity for l-lysine on the fish enzymes.
Collapse
Affiliation(s)
- Yasushi Tani
- College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; R-GIRO, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; Mitsubishi Chemical Group Science and Technology Research Center, Inc., Yokohama, Kanagawa 227-8502, Japan; and API Corporation, Yokohama, Kanagawa 227-8502, Japan College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; R-GIRO, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; Mitsubishi Chemical Group Science and Technology Research Center, Inc., Yokohama, Kanagawa 227-8502, Japan; and API Corporation, Yokohama, Kanagawa 227-8502, Japan
| | - Koichiro Omatsu
- College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; R-GIRO, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; Mitsubishi Chemical Group Science and Technology Research Center, Inc., Yokohama, Kanagawa 227-8502, Japan; and API Corporation, Yokohama, Kanagawa 227-8502, Japan
| | - Shigeki Saito
- College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; R-GIRO, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; Mitsubishi Chemical Group Science and Technology Research Center, Inc., Yokohama, Kanagawa 227-8502, Japan; and API Corporation, Yokohama, Kanagawa 227-8502, Japan College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; R-GIRO, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; Mitsubishi Chemical Group Science and Technology Research Center, Inc., Yokohama, Kanagawa 227-8502, Japan; and API Corporation, Yokohama, Kanagawa 227-8502, Japan
| | - Ryoma Miyake
- College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; R-GIRO, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; Mitsubishi Chemical Group Science and Technology Research Center, Inc., Yokohama, Kanagawa 227-8502, Japan; and API Corporation, Yokohama, Kanagawa 227-8502, Japan College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; R-GIRO, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; Mitsubishi Chemical Group Science and Technology Research Center, Inc., Yokohama, Kanagawa 227-8502, Japan; and API Corporation, Yokohama, Kanagawa 227-8502, Japan
| | - Hiroshi Kawabata
- College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; R-GIRO, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; Mitsubishi Chemical Group Science and Technology Research Center, Inc., Yokohama, Kanagawa 227-8502, Japan; and API Corporation, Yokohama, Kanagawa 227-8502, Japan College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; R-GIRO, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; Mitsubishi Chemical Group Science and Technology Research Center, Inc., Yokohama, Kanagawa 227-8502, Japan; and API Corporation, Yokohama, Kanagawa 227-8502, Japan
| | - Makoto Ueda
- College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; R-GIRO, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; Mitsubishi Chemical Group Science and Technology Research Center, Inc., Yokohama, Kanagawa 227-8502, Japan; and API Corporation, Yokohama, Kanagawa 227-8502, Japan
| | - Hisaaki Mihara
- College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; R-GIRO, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan; Mitsubishi Chemical Group Science and Technology Research Center, Inc., Yokohama, Kanagawa 227-8502, Japan; and API Corporation, Yokohama, Kanagawa 227-8502, Japan
| |
Collapse
|
82
|
Brahma RK, McCleary RJR, Kini RM, Doley R. Venom gland transcriptomics for identifying, cataloging, and characterizing venom proteins in snakes. Toxicon 2014; 93:1-10. [PMID: 25448392 DOI: 10.1016/j.toxicon.2014.10.022] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 10/27/2014] [Indexed: 01/13/2023]
Abstract
Snake venoms are cocktails of protein toxins that play important roles in capture and digestion of prey. Significant qualitative and quantitative variation in snake venom composition has been observed among and within species. Understanding these variations in protein components is instrumental in interpreting clinical symptoms during human envenomation and in searching for novel venom proteins with potential therapeutic applications. In the last decade, transcriptomic analyses of venom glands have helped in understanding the composition of various snake venoms in great detail. Here we review transcriptomic analysis as a powerful tool for understanding venom profile, variation and evolution.
Collapse
Affiliation(s)
- Rajeev Kungur Brahma
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784 028, Assam, India
| | - Ryan J R McCleary
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - R Manjunatha Kini
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore; Department of Biochemistry, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA; University of South Australia, School of Pharmacy and Medical Sciences, Adelaide, South Australia 5001, Australia
| | - Robin Doley
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784 028, Assam, India.
| |
Collapse
|
83
|
Abdelkafi-Koubaa Z, Jebali J, Othman H, Morjen M, Aissa I, Zouari-Kesentini R, Bazaa A, Ellefi AA, Majdoub H, Srairi-Abid N, Gargouri Y, El Ayeb M, Marrakchi N. A thermoactive l-amino acid oxidase from Cerastes cerastes snake venom: Purification, biochemical and molecular characterization. Toxicon 2014; 89:32-44. [DOI: 10.1016/j.toxicon.2014.06.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 06/14/2014] [Accepted: 06/25/2014] [Indexed: 11/28/2022]
|
84
|
Kopacz MM, Heuts DPHM, Fraaije MW. Kinetic mechanism of putrescine oxidase from Rhodococcus erythropolis. FEBS J 2014; 281:4384-93. [PMID: 25060191 DOI: 10.1111/febs.12945] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 07/01/2014] [Accepted: 07/23/2014] [Indexed: 01/14/2023]
Abstract
Putrescine oxidase from Rhodococcus erythropolis (PuO) is a flavin-containing amine oxidase from the monoamine oxidase family that performs oxidative deamination of aliphatic diamines. In this study we report pre-steady-state kinetic analyses of the enzyme with the use of single- and double-mixing stopped-flow spectroscopy and putrescine as a substrate. During the fast and irreversible reductive half-reaction no radical intermediates were observed, suggesting a direct hydride transfer from the substrate to the FAD. The rate constant of flavin reoxidation depends on the ligand binding; when the imine product was bound to the enzyme the rate constant was higher than with free enzyme species. Similar results were obtained with product-mimicking ligands and this indicates that a ternary complex is formed during catalysis. The obtained kinetic data were used together with steady-state rate equations derived for ping-pong, ordered sequential and bifurcated mechanisms to explore which mechanism is operative. The integrated analysis revealed that PuO employs a bifurcated mechanism due to comparable rate constants of product release from the reduced enzyme and reoxidation of the reduced enzyme-product complex.
Collapse
Affiliation(s)
- Malgorzata M Kopacz
- Molecular Enzymology Group, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, The Netherlands
| | | | | |
Collapse
|
85
|
Pollegioni L, Motta P, Molla G. L-amino acid oxidase as biocatalyst: a dream too far? Appl Microbiol Biotechnol 2014; 97:9323-41. [PMID: 24077723 DOI: 10.1007/s00253-013-5230-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 08/29/2013] [Accepted: 09/02/2013] [Indexed: 12/27/2022]
Abstract
L-amino acid oxidase (LAAO) is a flavoenzyme containing non-covalently bound flavin adenine dinucleotide, which catalyzes the stereospecific oxidative deamination of l-amino acids to α-keto acids and also produces ammonia and hydrogen peroxide via an imino acid intermediate. LAAOs purified from snake venoms are the best-studied members of this family of enzymes, although a number of LAAOs from bacterial and fungal sources have been also reported. From a biochemical point of view, LAAOs from different sources are distinguished by molecular mass, substrate specificity, post-translational modifications and regulation. In analogy to the well-known biotechnological applications of d-amino acid oxidase, important results are expected from the availability of suitable LAAOs; however, these expectations have not been fulfilled yet because none of the "true" LAAOs has successfully been expressed as a recombinant protein in prokaryotic hosts, such as Escherichia coli. In enzyme biotechnology, recombinant production of a protein is mandatory both for the production of large amounts of the catalyst and to improve its biochemical properties by protein engineering. As an alternative, flavoenzymes active on specific l-amino acids have been identified, e.g., l-aspartate oxidase, l-lysine oxidase, l-phenylalanine oxidase, etc. According to presently available information, amino acid oxidases with "narrow" or "strict" substrate specificity represent as good candidates to obtain an enzyme more suitable for biotechnological applications by enlarging their substrate specificity by means of protein engineering.
Collapse
|
86
|
Roberts KM, Tormos JR, Fitzpatrick PF. Characterization of unstable products of flavin- and pterin-dependent enzymes by continuous-flow mass spectrometry. Biochemistry 2014; 53:2672-9. [PMID: 24713088 PMCID: PMC4010283 DOI: 10.1021/bi500267c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
![]()
Continuous-flow mass spectrometry
(CFMS) was used to monitor the
products formed during the initial 0.25–20 s of the reactions
catalyzed by the flavoprotein N-acetylpolyamine oxidase
(PAO) and the pterin-dependent enzymes phenylalanine hydroxylase (PheH)
and tyrosine hydroxylase (TyrH). N,N′-Dibenzyl-1,4-diaminobutane (DBDB) is a substrate for PAO
for which amine oxidation is rate-limiting. CFMS of the reaction showed
formation of an initial imine due to oxidation of an exo-carbon–nitrogen bond. Nonenzymatic hydrolysis of the imine
formed benzaldehyde and N-benzyl-1,4-diaminobutane;
the subsequent oxidation by PAO of the latter to an additional imine
could also be followed. Measurement of the deuterium kinetic isotope
effect on DBDB oxidation by CFMS yielded a value of 7.6 ± 0.3,
in good agreement with a value of 6.7 ± 0.6 from steady-state
kinetic analyses. In the PheH reaction, the transient formation of
the 4a-hydroxypterin product was readily detected; tandem mass spectrometry
confirmed attachment of the oxygen to C(4a). With wild-type TyrH,
the 4a-hydroxypterin was also the product. In contrast, no product
other than a dihydropterin could be detected in the reaction of the
mutant protein E332A TyrH.
Collapse
Affiliation(s)
- Kenneth M Roberts
- Department of Biochemistry, University of Texas Health Science Center , San Antonio, Texas 78229, United States
| | | | | |
Collapse
|
87
|
Izidoro LFM, Sobrinho JC, Mendes MM, Costa TR, Grabner AN, Rodrigues VM, da Silva SL, Zanchi FB, Zuliani JP, Fernandes CFC, Calderon LA, Stábeli RG, Soares AM. Snake venom L-amino acid oxidases: trends in pharmacology and biochemistry. BIOMED RESEARCH INTERNATIONAL 2014; 2014:196754. [PMID: 24738050 PMCID: PMC3971498 DOI: 10.1155/2014/196754] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 12/13/2013] [Accepted: 12/16/2013] [Indexed: 11/26/2022]
Abstract
L-amino acid oxidases are enzymes found in several organisms, including venoms of snakes, where they contribute to the toxicity of ophidian envenomation. Their toxicity is primarily due to enzymatic activity, but other mechanisms have been proposed recently which require further investigation. L-amino acid oxidases exert biological and pharmacological effects, including actions on platelet aggregation and the induction of apoptosis, hemorrhage, and cytotoxicity. These proteins present a high biotechnological potential for the development of antimicrobial, antitumor, and antiprotozoan agents. This review provides an overview of the biochemical properties and pharmacological effects of snake venom L-amino acid oxidases, their structure/activity relationship, and supposed mechanisms of action described so far.
Collapse
Affiliation(s)
- Luiz Fernando M. Izidoro
- Faculdade de Ciências Integradas do Pontal e Departamento de Genética e Bioquímica, Universidade Federal de Uberlândia (UFU), Uberlândia, MG, Brazil
| | - Juliana C. Sobrinho
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, (CEBio), Fundação Oswaldo Cruz, Fiocruz Rondônia e Departamento de Medicina, Universidade Federal de Rondônia (UNIR), Porto Velho, RO, Brazil
| | - Mirian M. Mendes
- Faculdade de Ciências Integradas do Pontal e Departamento de Genética e Bioquímica, Universidade Federal de Uberlândia (UFU), Uberlândia, MG, Brazil
| | - Tássia R. Costa
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto (FCFRP), Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Amy N. Grabner
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, (CEBio), Fundação Oswaldo Cruz, Fiocruz Rondônia e Departamento de Medicina, Universidade Federal de Rondônia (UNIR), Porto Velho, RO, Brazil
| | - Veridiana M. Rodrigues
- Faculdade de Ciências Integradas do Pontal e Departamento de Genética e Bioquímica, Universidade Federal de Uberlândia (UFU), Uberlândia, MG, Brazil
| | - Saulo L. da Silva
- Departamento de Química, Biotecnologia e Engenharia de Bioprocessos, Universidade Federal de São João del Rei (UFSJ), Campus Altoparaopeba, Ouro Branco, MG, Brazil
| | - Fernando B. Zanchi
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, (CEBio), Fundação Oswaldo Cruz, Fiocruz Rondônia e Departamento de Medicina, Universidade Federal de Rondônia (UNIR), Porto Velho, RO, Brazil
| | - Juliana P. Zuliani
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, (CEBio), Fundação Oswaldo Cruz, Fiocruz Rondônia e Departamento de Medicina, Universidade Federal de Rondônia (UNIR), Porto Velho, RO, Brazil
| | - Carla F. C. Fernandes
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, (CEBio), Fundação Oswaldo Cruz, Fiocruz Rondônia e Departamento de Medicina, Universidade Federal de Rondônia (UNIR), Porto Velho, RO, Brazil
| | - Leonardo A. Calderon
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, (CEBio), Fundação Oswaldo Cruz, Fiocruz Rondônia e Departamento de Medicina, Universidade Federal de Rondônia (UNIR), Porto Velho, RO, Brazil
| | - Rodrigo G. Stábeli
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, (CEBio), Fundação Oswaldo Cruz, Fiocruz Rondônia e Departamento de Medicina, Universidade Federal de Rondônia (UNIR), Porto Velho, RO, Brazil
| | - Andreimar M. Soares
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, (CEBio), Fundação Oswaldo Cruz, Fiocruz Rondônia e Departamento de Medicina, Universidade Federal de Rondônia (UNIR), Porto Velho, RO, Brazil
| |
Collapse
|
88
|
Matsui D, Im DH, Sugawara A, Fukuta Y, Fushinobu S, Isobe K, Asano Y. Mutational and crystallographic analysis of l-amino acid oxidase/monooxygenase from Pseudomonas sp. AIU 813: Interconversion between oxidase and monooxygenase activities. FEBS Open Bio 2014; 4:220-8. [PMID: 24693490 PMCID: PMC3970082 DOI: 10.1016/j.fob.2014.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 01/31/2014] [Accepted: 02/03/2014] [Indexed: 10/31/2022] Open
Abstract
In this study, it was shown for the first time that l-amino acid oxidase of Pseudomonas sp. AIU813, renamed as l-amino acid oxidase/monooxygenase (l-AAO/MOG), exhibits l-lysine 2-monooxygenase as well as oxidase activity. l-Lysine oxidase activity of l-AAO/MOG was increased in a p-chloromercuribenzoate (p-CMB) concentration-dependent manner to a final level that was five fold higher than that of the non-treated enzyme. In order to explain the effects of modification by the sulfhydryl reagent, saturation mutagenesis studies were carried out on five cysteine residues, and we succeeded in identifying l-AAO/MOG C254I mutant enzyme, which showed five-times higher specific activity of oxidase activity than that of wild type. The monooxygenase activity shown by the C254I variant was decreased significantly. Moreover, we also determined a high-resolution three-dimensional structure of l-AAO/MOG to provide a structural basis for its biochemical characteristics. The key residue for the activity conversion of l-AAO/MOG, Cys-254, is located near the aromatic cage (Trp-418, Phe-473, and Trp-516). Although the location of Cys-254 indicates that it is not directly involved in the substrate binding, the chemical modification by p-CMB or C254I mutation would have a significant impact on the substrate binding via the side chain of Trp-516. It is suggested that a slight difference of the binding position of a substrate can dictate the activity of this type of enzyme as oxidase or monooxygenase.
Collapse
Key Words
- 4-AA, 4-aminoantipyrine
- CHCA, α-Cyano-4-hydroxycinnamic acid
- Crystallography
- FMOs, flavin monooxygenases
- Flavin monooxygenases
- Flavin-containing monoamine oxidase family
- LB, Luria–Bertani
- LGOX, l-glutamate oxidase
- MAO, flavin-containing monoamine oxidase
- PAO, l-phenylalanine oxidase
- Saturation mutagenesis
- TFA, trifluoroacetic acid
- TMO, l-tryptophan 2-monooxygenase
- TOOS, N-ethyl-N-(2-hydroxy-3-sulfopropyl)-3-methylaniline
- amid, amide hydrolase gene
- l-AAO, l-amino acid oxidase
- l-AAO/MOG, l-amino acid oxidase/monooxygenase
- l-Amino acid oxidase/monooxygenase
- laao/mog, l-amino acid oxidase/monooxygenase gene
- p-CMB, p-chloromercuribenzoate
Collapse
Affiliation(s)
- Daisuke Matsui
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan ; Asano Active Enzyme Molecule Project, ERATO, JST, 5180 Kurokawa, Imizu,Toyama 939-0398, Japan
| | - Do-Hyun Im
- Department of Biotechnology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Asami Sugawara
- Department of Biological Chemistry and Food Science, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka 020-8550, Japan
| | - Yasuhisa Fukuta
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| | - Shinya Fushinobu
- Asano Active Enzyme Molecule Project, ERATO, JST, 5180 Kurokawa, Imizu,Toyama 939-0398, Japan ; Department of Biotechnology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kimiyasu Isobe
- Department of Biological Chemistry and Food Science, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka 020-8550, Japan
| | - Yasuhisa Asano
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan ; Asano Active Enzyme Molecule Project, ERATO, JST, 5180 Kurokawa, Imizu,Toyama 939-0398, Japan
| |
Collapse
|
89
|
Effect of L-amino acid oxidase from Calloselasma rhodosthoma snake venom on human neutrophils. Toxicon 2014; 80:27-37. [PMID: 24462716 DOI: 10.1016/j.toxicon.2013.12.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 12/02/2013] [Accepted: 12/30/2013] [Indexed: 01/01/2023]
Abstract
The in vitro effects of LAAO, an l-amino acid oxidase isolated from Calloselasma rhodosthoma snake venom, on isolated human neutrophil function were investigated. LAAO showed no toxicity on neutrophils. At non-cytotoxic concentrations, LAAO induced the superoxide anion production by isolated human neutrophil. This toxin, in its native form, is also able to stimulate the production of hydrogen peroxide in neutrophils, suggesting that its primary structure is essential for stimulation the cell. Moreover, the incubation of LAAO and phenol red medium did not induce the production of hydrogen peroxide. Furthermore, LAAO was able to stimulate neutrophils to release proinflammatory mediators such as IL-8 and TNF-α as well as NETs liberation. Together, the data showed that the LAAO triggers relevant proinflammatory events. Particular regions of the molecule distinct from the LAAO catalytic site may be involved in the onset of inflammatory events.
Collapse
|
90
|
L-Amino acid oxidases from microbial sources: types, properties, functions, and applications. Appl Microbiol Biotechnol 2013; 98:1507-15. [PMID: 24352734 DOI: 10.1007/s00253-013-5444-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 11/26/2013] [Accepted: 11/27/2013] [Indexed: 10/25/2022]
Abstract
L-Amino acid oxidases (LAAOs), which catalyze the stereospecific oxidative deamination of L-amino acids to α-keto acids and ammonia, are flavin adenine dinucleotide-containing homodimeric proteins. L-Amino acid oxidases are widely distributed in diverse organisms and have a range of properties. Because expressing LAAOs as recombinant proteins in heterologous hosts is difficult, their biotechnological applications have not been thoroughly advanced. LAAOs are thought to contribute to amino acid catabolism, enhance iron acquisition, display antimicrobial activity, and catalyze keto acid production, among other roles. Here, we review the types, properties, structures, biological functions, heterologous expression, and applications of LAAOs obtained from microbial sources. We expect this review to increase interest in LAAO studies.
Collapse
|
91
|
Beaupre BA, Carmichael BR, Hoag MR, Shah DD, Moran GR. Renalase is an α-NAD(P)H oxidase/anomerase. J Am Chem Soc 2013; 135:13980-7. [PMID: 23964689 DOI: 10.1021/ja407384h] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Renalase is a protein hormone secreted into the blood by the kidney that is reported to lower blood pressure and slow heart rate. Since its discovery in 2005, renalase has been the subject of conjecture pertaining to its catalytic function. While it has been widely reported that renalase is the third monoamine oxidase (monoamine oxidase C) that oxidizes circulating catecholamines such as epinephrine, there has been no convincing demonstration of this catalysis in vitro. Renalase is a flavoprotein whose structural topology is similar to known oxidases, lysine demethylases, and monooxygenases, but its active site bears no resemblance to that of any known flavoprotein. We have identified the catalytic activity of renalase as an α-NAD(P)H oxidase/anomerase, whereby low equilibrium concentrations of the α-anomer of NADPH and NADH initiate rapid reduction of the renalase flavin cofactor. The reduced cofactor then reacts with dioxygen to form hydrogen peroxide and releases nicotinamide dinucleotide product in the β-form. These processes yield an apparent turnover number (0.5 s(-1) in atmospheric dioxygen) that is at least 2 orders of magnitude more rapid than any reported activity with catechol neurotransmitters. This highly novel activity is the first demonstration of a role for naturally occurring α-NAD(P)H anomers in mammalian physiology and the first report of a flavoprotein catalyzing an epimerization reaction.
Collapse
Affiliation(s)
- Brett A Beaupre
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee , 3210 N. Cramer Street, Milwaukee, Wisconsin 53211-3209, United States
| | | | | | | | | |
Collapse
|
92
|
Affiliation(s)
- Artur Gora
- Loschmidt Laboratories,
Department
of Experimental Biology and Research Centre for Toxic Compounds in
the Environment, Faculty of Science, Masaryk University, Kamenice 5/A13, 625 00 Brno, Czech Republic
| | - Jan Brezovsky
- Loschmidt Laboratories,
Department
of Experimental Biology and Research Centre for Toxic Compounds in
the Environment, Faculty of Science, Masaryk University, Kamenice 5/A13, 625 00 Brno, Czech Republic
| | - Jiri Damborsky
- Loschmidt Laboratories,
Department
of Experimental Biology and Research Centre for Toxic Compounds in
the Environment, Faculty of Science, Masaryk University, Kamenice 5/A13, 625 00 Brno, Czech Republic
- International Centre for Clinical
Research, St. Anne’s University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| |
Collapse
|
93
|
Abstract
Since cancer is one of the leading causes of death worldwide, and there is an urgent need to find better treatment. In recent years remarkable progress has been made towards the understanding of proposed hallmarks of cancer development and treatment. Treatment modalities comprise radiation therapy, surgery, chemotherapy, immunotherapy and hormonal therapy. Currently, the use of chemotherapeutics remains the predominant option for clinical control. However, one of the major problems with successful cancer therapy using chemotherapeutics is that patients often do not respond or eventually develop resistance after initial treatment. This has led to the increased use of anticancer drugs developed from natural resources. The biodiversity of venoms and toxins makes them a unique source from which novel therapeutics may be developed. In this review, the anticancer potential of snake venom is discussed. Some of the included molecules are under clinical trial and may find application for anticancer drug development in the near future.
Collapse
Affiliation(s)
- Deepika Jain
- IGNOU-I2IT Centre of Excellence for Advanced Education and Research, Pune, Maharashtra, India
| | | |
Collapse
|
94
|
Gaweska HM, Taylor AB, Hart PJ, Fitzpatrick PF. Structure of the flavoprotein tryptophan 2-monooxygenase, a key enzyme in the formation of galls in plants. Biochemistry 2013; 52:2620-6. [PMID: 23521653 DOI: 10.1021/bi4001563] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The flavoprotein tryptophan 2-monooxygenase catalyzes the oxidative decarboxylation of tryptophan to yield indole-3-acetamide. This is the initial step in the biosynthesis of the plant growth hormone indole-acetic acid by bacterial pathogens that cause crown gall and related diseases. The structure of the enzyme from Pseudomonas savastanoi has been determined by X-ray diffraction methods to a resolution of 1.95 Å. The overall structure of the protein shows that it has the same fold as members of the monoamine oxidase family of flavoproteins, with the greatest similarities to the l-amino acid oxidases. The location of bound indole-3-acetamide in the active site allows identification of residues responsible for substrate binding and specificity. Two residues in the enzyme are conserved in all members of the monoamine oxidase family, Lys365 and Trp466. The K365M mutation decreases the kcat and kcat/KTrp values by 60000- and 2 million-fold, respectively. The deuterium kinetic isotope effect increases to 3.2, consistent with carbon-hydrogen bond cleavage becoming rate-limiting in the mutant enzyme. The W466F mutation decreases the kcat value <2-fold and the kcat/KTrp value only 5-fold, while the W466M mutation results in an enzyme lacking flavin and detectable activity. This is consistent with a role for Trp466 in maintaining the structure of the flavin-binding site in the more conserved FAD domain.
Collapse
Affiliation(s)
- Helena M Gaweska
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, Texas 78229, USA
| | | | | | | |
Collapse
|
95
|
Volkov A, Khoshnevis S, Neumann P, Herrfurth C, Wohlwend D, Ficner R, Feussner I. Crystal structure analysis of a fatty acid double-bond hydratase from Lactobacillus acidophilus. ACTA CRYSTALLOGRAPHICA SECTION D: BIOLOGICAL CRYSTALLOGRAPHY 2013; 69:648-57. [PMID: 23519674 DOI: 10.1107/s0907444913000991] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 01/10/2013] [Indexed: 11/10/2022]
Abstract
Bacteria have evolved mechanisms for the hydrogenation of unsaturated fatty acids. Hydroxy fatty acid formation may be the first step in such a process; however, knowledge of the structural and mechanistic aspects of this reaction is scarce. Recently, myosin cross-reactive antigen was shown to be a bacterial FAD-containing hydratase which acts on the 9Z and 12Z double bonds of C16 and C18 non-esterified fatty acids, with the formation of 10-hydroxy and 10,13-dihydroxy fatty acids. These fatty acid hydratases form a large protein family which is conserved across Gram-positive and Gram-negative bacteria with no sequence similarity to any known protein apart from the FAD-binding motif. In order to shed light on the substrate recognition and the mechanism of the hydratase reaction, the crystal structure of the hydratase from Lactobacillus acidophilus (LAH) was determined by single-wavelength anomalous dispersion. Crystal structures of apo LAH and of LAH with bound linoleic acid were refined at resolutions of 2.3 and 1.8 Å, respectively. LAH is a homodimer; each protomer consists of four intricately connected domains. Three of them form the FAD-binding and substrate-binding sites and reveal structural similarity to three domains of several flavin-dependent enzymes, including amine oxidoreductases. The additional fourth domain of LAH is located at the C-terminus and consists of three α-helices. It covers the entrance to the hydrophobic substrate channel leading from the protein surface to the active site. In the presence of linoleic acid, the fourth domain of one protomer undergoes conformational changes and opens the entrance to the substrate-binding channel of the other protomer of the LAH homodimer. The linoleic acid molecule is bound at the entrance to the substrate channel, suggesting movement of the lid domain triggered by substrate recognition.
Collapse
Affiliation(s)
- Anton Volkov
- Department for Plant Biochemistry, Albrecht-von-Haller-Institute for Plant Sciences, Georg-August-University, Göttingen, Germany
| | | | | | | | | | | | | |
Collapse
|
96
|
Mitra J, Bhattacharyya D. Irreversible inactivation of snake venom l-amino acid oxidase by covalent modification during catalysis of l-propargylglycine. FEBS Open Bio 2013; 3:135-43. [PMID: 23772385 PMCID: PMC3668516 DOI: 10.1016/j.fob.2013.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 11/18/2022] Open
Abstract
Snake venom l-amino acid oxidase (SV-LAAO, a flavor-enzyme) has attracted considerable attention due to its multifunctional nature, which is manifest in diverse clinical and biological effects such as inhibition of platelet aggregation, induction of cell apoptosis and cytotoxicity against various cells. The majority of these effects are mediated by H2O2 generated during the catalytic conversion of l-amino acids. The substrate analog l-propargylglycine (LPG) irreversibly inhibited the enzyme from Crotalus adamanteus and Crotalus atrox in a dose- and time-dependent manner. Inactivation was irreversible which was significantly protected by the substrate l-phenylalanine. A Kitz-Wilson replot of the inhibition kinetics suggested formation of reversible enzyme-LPG complex, which occurred prior to modification and inactivation of the enzyme. UV-visible and fluorescence spectra of the enzyme and the cofactor strongly suggested formation of covalent adduct between LPG and an active site residue of the enzyme. A molecular modeling study revealed that the FAD-binding, substrate-binding and the helical domains are conserved in SV-LAAOs and both His223 and Arg322 are the important active site residues that are likely to get modified by LPG. Chymotrypsin digest of the LPG inactivated enzyme followed by RP-HPLC and MALDI mass analysis identified His223 as the site of modification. The findings reported here contribute towards complete inactivation of SV-LAAO as a part of snake envenomation management.
Collapse
Key Words
- CHD, 1,2-cyclohexanedione
- Crotalus adamanteus venom
- Crotalus atrox venom
- DEPC, diethylpyrocarbonate
- FAD, flavin adenine dinucleotide
- Gdn-HCl, guanidine hydrochloride
- Irreversible inactivation
- LAAO, l-amino acid oxidase (EC. 1.4.3.2)
- LPG, l-propargylglycine
- MALDI-TOF, matrix-assisted laser desorption ionization-time of flight
- Mechanism-based inhibitor
- TNBS, trinitrobenzene sulfonic acid.
- l-Amino acid oxidase
- l-Phe, l-phenylalaine
- l-Propargylglycine
Collapse
|
97
|
A thermostable L-aspartate oxidase: a new tool for biotechnological applications. Appl Microbiol Biotechnol 2013; 97:7285-95. [PMID: 23371294 DOI: 10.1007/s00253-013-4688-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 12/22/2012] [Accepted: 12/27/2012] [Indexed: 11/27/2022]
Abstract
L-Amino acid oxidases (LAAOs) are homodimeric flavin adenine dinucleotide (FAD)-containing flavoproteins that catalyze the stereospecific oxidative deamination of L-amino acids to α-keto acids, ammonia, and hydrogen peroxide. Unlike the D-selective counterpart, the biotechnological application of LAAOs has not been thoroughly advanced because of the difficulties in their expression as recombinant protein in prokaryotic hosts. In this work, L-aspartate oxidase from the thermophilic archea Sulfolobus tokodaii (StLASPO, specific for L-aspartate and L-asparagine only) was efficiently produced as recombinant protein in E. coli in the active form as holoenzyme. This recombinant flavoenzyme shows the classical properties of FAD-containing oxidases. Indeed, StLASPO shows distinctive features that makes it attractive for biotechnological applications: high thermal stability (it is fully stable up to 80 °C) and high temperature optimum, stable activity in a broad range of pH (7.0-10.0), weak inhibition by the product oxaloacetate and by D-aspartate, and tight binding of the FAD cofactor. This latter property significantly distinguishes StLASPO from the E. coli counterpart. StLASPO represents an appropriate novel biocatalyst for the production of D-aspartate and a well-suited protein scaffold to evolve a LAAO activity by protein engineering.
Collapse
|
98
|
Cloning of a novel nicotine oxidase gene from Pseudomonas sp. strain HZN6 whose product nonenantioselectively degrades nicotine to pseudooxynicotine. Appl Environ Microbiol 2013; 79:2164-71. [PMID: 23335761 DOI: 10.1128/aem.03824-12] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas sp. strain HZN6 utilizes nicotine as its sole source of carbon, nitrogen, and energy. However, its catabolic mechanism has not been elucidated. In this study, self-formed adaptor PCR was performed to amplify the upstream sequence of the pseudooxynicotine amine oxidase gene. A 1,437-bp open reading frame (designated nox) was found to encode a nicotine oxidase (NOX) that shows 30% amino acid sequence identity with 6-hydroxy-l-nicotine oxidase from Arthrobacter nicotinovorans. The nox gene was cloned into a broad-host-range cloning vector and transferred into the non-nicotine-degrading bacteria Escherichia coli DH5α (DH-nox) and Pseudomonas putida KT2440 (KT-nox). The transconjugant KT-nox obtained nicotine degradation ability and yielded an equimolar amount of pseudooxynicotine, while DH-nox did not. Reverse transcription-PCR showed that the nox gene is expressed in both DH5α and KT2440, suggesting that additional factors required for nicotine degradation are present in a Pseudomonas strain(s), but not in E. coli. The mutant of strain HZN6 with nox disrupted lost the ability to degrade nicotine, but not pseudooxynicotine. These results suggested that the nox gene is responsible for the first step of nicotine degradation. The (RS)-nicotine degradation results showed that the two enantiomers were degraded at approximately the same rate, indicating that NOX does not show chiral selectivity. Site-directed mutagenesis revealed that both the conserved flavin adenine dinucleotide (FAD)-binding GXGXXG motif and His456 are essential for nicotine degradation activity.
Collapse
|
99
|
Bhattacharjee P, Bhattacharyya D. Characterization of the aqueous extract of the root of Aristolochia indica: evaluation of its traditional use as an antidote for snake bites. JOURNAL OF ETHNOPHARMACOLOGY 2013; 145:220-6. [PMID: 23174522 DOI: 10.1016/j.jep.2012.10.056] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 10/17/2012] [Accepted: 10/30/2012] [Indexed: 06/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The aqueous extract of the roots of Aristolochia indica is used as a decoction for the ailment of a number of diseases including snake bite treatment. Though the alcoholic extract of the different parts of the plant are well studied, information on the aqueous extract is limited. We have estimated aristolochic acid, different enzymes, enzyme inhibitors and anti-snake venom potency of its root extract. MATERIALS AND METHODS Reverse phase-HPLC was used to quantify aristolochic acid. Zymography, DQ-gelatin assay and atomic force microscopy were done to demonstrate gelatinase and collagenase activities of the extract. SDS-PAGE followed by MS/MS analysis revealed the identity of major protein components. Toxicity of the extract was estimated on animal model. Interaction of the extract with Russell's viper venom components was followed by Rayleigh scattering and enzyme assay. RESULTS The aristolochic acid content of the root extract is 3.08 ± 1.88 × 10(-3)mg/ml. The extract possesses strong gelatinolytic, collagenase, peroxidase and nuclease activities together with l-amino acid oxidase and protease inhibitory potencies. Partial proteomic studies indicated presence of starch branching enzymes as major protein constituent of the extract. The extract did not show any acute and sub-chronic toxicity in animals at lower doses, but high dose causes liver and kidney damage. The extract elongated duration of survival of animals after application of Russell's viper venom. CONCLUSIONS Considering the low aristolochic acid content of the extract, its consumption for a short time at moderate dose does not appear to cause serious toxicity. Strong inhibition of l-amino acid oxidase may give partial relief from snake bite after topical application of the extract.
Collapse
Affiliation(s)
- Payel Bhattacharjee
- Division of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | | |
Collapse
|
100
|
Isolation and biochemical, functional and structural characterization of a novel l-amino acid oxidase from Lachesis muta snake venom. Toxicon 2012; 60:1263-76. [DOI: 10.1016/j.toxicon.2012.08.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 08/06/2012] [Accepted: 08/09/2012] [Indexed: 11/20/2022]
|