Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 296
Filter
Add more filters

Publication year range
1.
Cell ; 184(6): 1604-1620, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33740455

ABSTRACT

Historically, emerging viruses appear constantly and have cost millions of human lives. Currently, climate change and intense globalization have created favorable conditions for viral transmission. Therefore, effective antivirals, especially those targeting the conserved protein in multiple unrelated viruses, such as the compounds targeting RNA-dependent RNA polymerase, are urgently needed to combat more emerging and re-emerging viruses in the future. Here we reviewed the development of antivirals with common targets, including those against the same protein across viruses, or the same viral function, to provide clues for development of antivirals for future epidemics.


Subject(s)
Antiviral Agents/therapeutic use , Communicable Diseases, Emerging/drug therapy , Communicable Diseases, Emerging/epidemiology , Molecular Targeted Therapy/methods , Pandemics , Virus Diseases/drug therapy , Virus Diseases/epidemiology , Viruses/enzymology , Animals , Antiviral Agents/pharmacology , Communicable Diseases, Emerging/virology , Humans , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Envelope Proteins/antagonists & inhibitors , Virus Diseases/virology , Virus Internalization/drug effects
2.
Nature ; 577(7791): 572-575, 2020 01.
Article in English | MEDLINE | ID: mdl-31942067

ABSTRACT

The CRISPR system in bacteria and archaea provides adaptive immunity against mobile genetic elements. Type III CRISPR systems detect viral RNA, resulting in the activation of two regions of the Cas10 protein: an HD nuclease domain (which degrades viral DNA)1,2 and a cyclase domain (which synthesizes cyclic oligoadenylates from ATP)3-5. Cyclic oligoadenylates in turn activate defence enzymes with a CRISPR-associated Rossmann fold domain6, sculpting a powerful antiviral response7-10 that can drive viruses to extinction7,8. Cyclic nucleotides are increasingly implicated in host-pathogen interactions11-13. Here we identify a new family of viral anti-CRISPR (Acr) enzymes that rapidly degrade cyclic tetra-adenylate (cA4). The viral ring nuclease AcrIII-1 is widely distributed in archaeal and bacterial viruses and in proviruses. The enzyme uses a previously unknown fold to bind cA4 specifically, and a conserved active site to rapidly cleave this signalling molecule, allowing viruses to neutralize the type III CRISPR defence system. The AcrIII-1 family has a broad host range, as it targets cA4 signalling molecules rather than specific CRISPR effector proteins. Our findings highlight the crucial role of cyclic nucleotide signalling in the conflict between viruses and their hosts.


Subject(s)
CRISPR-Cas Systems/immunology , Endonucleases/metabolism , Host Microbial Interactions/immunology , Sulfolobus/virology , Viral Proteins/metabolism , Viruses/enzymology , Adenine Nucleotides/chemistry , Adenine Nucleotides/metabolism , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/metabolism , DNA, Viral/metabolism , Endonucleases/chemistry , Models, Molecular , Nucleotides, Cyclic/chemistry , Nucleotides, Cyclic/metabolism , Oligoribonucleotides/chemistry , Oligoribonucleotides/metabolism , Phylogeny , Signal Transduction , Sulfolobus/genetics , Sulfolobus/immunology , Sulfolobus/metabolism , Viral Proteins/chemistry , Viral Proteins/classification , Viruses/immunology
3.
Proc Natl Acad Sci U S A ; 116(25): 12343-12352, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31167942

ABSTRACT

Genes encoding cytochrome P450 (CYP; P450) enzymes occur widely in the Archaea, Bacteria, and Eukarya, where they play important roles in metabolism of endogenous regulatory molecules and exogenous chemicals. We now report that genes for multiple and unique P450s occur commonly in giant viruses in the Mimiviridae, Pandoraviridae, and other families in the proposed order Megavirales. P450 genes were also identified in a herpesvirus (Ranid herpesvirus 3) and a phage (Mycobacterium phage Adler). The Adler phage P450 was classified as CYP102L1, and the crystal structure of the open form was solved at 2.5 Å. Genes encoding known redox partners for P450s (cytochrome P450 reductase, ferredoxin and ferredoxin reductase, and flavodoxin and flavodoxin reductase) were not found in any viral genome so far described, implying that host redox partners may drive viral P450 activities. Giant virus P450 proteins share no more than 25% identity with the P450 gene products we identified in Acanthamoeba castellanii, an amoeba host for many giant viruses. Thus, the origin of the unique P450 genes in giant viruses remains unknown. If giant virus P450 genes were acquired from a host, we suggest it could have been from an as yet unknown and possibly ancient host. These studies expand the horizon in the evolution and diversity of the enormously important P450 superfamily. Determining the origin and function of P450s in giant viruses may help to discern the origin of the giant viruses themselves.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Evolution, Molecular , Multigene Family , Viruses/enzymology , Cytochrome P-450 Enzyme System/genetics
4.
Int J Mol Sci ; 22(9)2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33922750

ABSTRACT

The ubiquitin (Ub) proteasome system (UPS) plays a pivotal role in regulation of numerous cellular processes, including innate and adaptive immune responses that are essential for restriction of the virus life cycle in the infected cells. Deubiquitination by the deubiquitinating enzyme, deubiquitinase (DUB), is a reversible molecular process to remove Ub or Ub chains from the target proteins. Deubiquitination is an integral strategy within the UPS in regulating survival and proliferation of the infecting virus and the virus-invaded cells. Many viruses in the infected cells are reported to encode viral DUB, and these vial DUBs actively disrupt cellular Ub-dependent processes to suppress host antiviral immune response, enhancing virus replication and thus proliferation. This review surveys the types of DUBs encoded by different viruses and their molecular processes for how the infecting viruses take advantage of the DUB system to evade the host immune response and expedite their replication.


Subject(s)
Deubiquitinating Enzymes/metabolism , Host-Pathogen Interactions/immunology , Immunity, Innate/immunology , Ubiquitin/metabolism , Viral Proteins/metabolism , Virus Diseases/immunology , Viruses/enzymology , Animals , Deubiquitinating Enzymes/chemistry , Humans , Immune Evasion , Life Cycle Stages , Ubiquitination , Viral Proteins/chemistry , Virus Diseases/enzymology , Virus Diseases/virology , Virus Replication , Viruses/immunology
5.
Curr Top Microbiol Immunol ; 420: 131-154, 2019.
Article in English | MEDLINE | ID: mdl-30244323

ABSTRACT

Successful viral infection, as well as any resultant antiviral response, relies on numerous sequential interactions between host and viral factors. These interactions can take the form of affinity-based interactions between viral and host macromolecules or active, enzyme-based interactions, consisting both of direct enzyme activity performed by viral enzymes and indirect modulation of the activity of the host cell's enzymes via viral interference. This activity has the potential to transform the local microenvironment to the benefit or detriment of both the virus and the host, favouring either the continuation of the viral life cycle or the host's antiviral response. Comprehensive characterisation of enzymatic activity during viral infection is therefore necessary for the understanding of virally induced diseases. Activity-based protein profiling techniques have been established as effective and practicable tools with which to interrogate the regulation of enzymes' catalytic activity and the roles played by these enzymes in various cell processes. This paper will review the contributions of these techniques in characterising the roles of both host and viral enzymes during viral infection in humans.


Subject(s)
Host-Pathogen Interactions/physiology , Proteome/analysis , Proteome/metabolism , Proteomics/methods , Virus Diseases/metabolism , Virus Diseases/virology , Viruses/metabolism , Antiviral Agents/metabolism , Humans , Proteome/chemistry , Virus Diseases/enzymology , Virus Replication , Viruses/enzymology
6.
Chem Rev ; 118(18): 8786-8797, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30133258

ABSTRACT

Enzymes catalyze a vast range of reactions. Their catalytic performances, mechanisms, global folds, and active-site architectures are also highly diverse, suggesting that enzymes are shaped by an entire range of physiological demands and evolutionary constraints, as well as by chemical and physicochemical constraints. We have attempted to identify signatures of these shaping demands and constraints. To this end, we describe a bird's-eye view of the enzyme space from two angles: evolution and chemistry. We examine various chemical reaction parameters that may have shaped the catalytic performances and active-site architectures of enzymes. We test and weigh these considerations against physiological and evolutionary factors. Although the catalytic properties of the "average" enzyme correlate with cellular metabolic demands and enzyme expression levels, at the level of individual enzymes, a multitude of physiological demands and constraints, combined with the coincidental nature of evolutionary processes, result in a complex picture. Indeed, neither reaction type (a chemical constraint) nor evolutionary origin alone can explain enzyme rates. Nevertheless, chemical constraints are apparent in the convergence of active-site architectures in independently evolved enzymes, although significant variations within an architecture are common.


Subject(s)
Enzymes/chemistry , Enzymes/physiology , Evolution, Molecular , Animals , Archaea/enzymology , Bacteria/enzymology , Catalysis , Catalytic Domain , Diffusion , Fungi/enzymology , Humans , Kinetics , Protein Conformation , Viruses/enzymology
8.
Nucleic Acids Res ; 46(5): 2521-2536, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29361162

ABSTRACT

Genomes of halophilic archaea typically contain multiple loci of integrated mobile genetic elements (MGEs). Despite the abundance of these elements, however, mechanisms underlying their site-specific integration and excision have not been investigated. Here, we identified and characterized a novel recombination system encoded by the temperate pleolipovirus SNJ2, which infects haloarchaeon Natrinema sp. J7-1. SNJ2 genome is inserted into the tRNAMet gene and flanked by 14 bp direct repeats corresponding to attachment core sites. We showed that SNJ2 encodes an integrase (IntSNJ2) that excises the proviral genome from its host cell chromosome, but requires two small accessory proteins, Orf2 and Orf3, for integration. These proteins were co-transcribed with IntSNJ2 to form an operon. Homology searches showed that IntSNJ2-type integrases are widespread in haloarchaeal genomes and are associated with various integrated MGEs. Importantly, we confirmed that SNJ2-like recombination systems are encoded by haloarchaea from three different genera and are critical for integration and excision. Finally, phylogenetic analysis suggested that IntSNJ2-type recombinases belong to a novel family of archaeal integrases distinct from previously characterized recombinases, including those from the archaeal SSV- and pNOB8-type families.


Subject(s)
Integrases/metabolism , Viruses/enzymology , Archaea/enzymology , Archaea/genetics , Chromosomes, Archaeal , Integrases/biosynthesis , Integrases/classification , Integrases/genetics , Interspersed Repetitive Sequences , Proviruses/physiology , Recombination, Genetic , Transcription, Genetic , Tyrosine , Viral Proteins/biosynthesis , Viral Proteins/genetics , Viral Proteins/physiology , Virus Integration , Viruses/genetics
9.
Molecules ; 25(15)2020 Jul 26.
Article in English | MEDLINE | ID: mdl-32722555

ABSTRACT

In this study, the chemical diversity of polyphenols in Iris lactea var. chinensis seeds was identified by combined MS/MS-NMR analysis. Based on the annotated chemical profile, the isolation of stilbene oligomers was conducted, and consequently, stilbene oligomers (1-10) were characterized. Of these, compounds 1 and 2 are previously undescribed stilbene dimer glycoside (1) and tetramer glycoside (2), respectively. Besides, to evaluate this plant seed as a rich source of stilbene oligomers, we quantified three stilbene oligomers of I. lactea var. chinensis seeds. The contents of three major stilbene oligomers-trans-ε-viniferin (3), vitisin A (6), and vitisin B (9)-in I. lactea var. chinensis seeds were quantified as 2.32 (3), 4.95 (6), and 1.64 (9) mg/g dry weight (DW). All the isolated compounds were tested for their inhibitory activities against influenza neuraminidase. Compound 10 was found to be active with the half maximal inhibitory concentration (IC50) values at 4.76 µM. Taken together, it is concluded that I. lactea var. chinensis seed is a valuable source of stilbene oligomers with a human health benefit.


Subject(s)
Iris Plant/chemistry , Neuraminidase/antagonists & inhibitors , Polyphenols/chemistry , Viruses/drug effects , Humans , Plant Roots/chemistry , Polyphenols/pharmacology , Seeds/chemistry , Tandem Mass Spectrometry , Viruses/enzymology
10.
Adv Exp Med Biol ; 1142: 115-129, 2019.
Article in English | MEDLINE | ID: mdl-31102244

ABSTRACT

Lytic polysaccharide monooxygenases (LPMOs) are copper-dependent enzymes that catalyze the cleavage of 1,4-glycosidic bonds various plant cell wall polysaccharides and chitin. In contrast to glycoside hydrolases, LPMOs are active on the crystalline regions of polysaccharides and thus synergize with hydrolytic enzymes. This synergism leads to an overall increase in the biomass-degradation activity of enzyme mixtures. Chitin-active LPMOs were discovered in 2010 and are currently classified in families AA10, AA11, and AA15 of the Carbohydrate-Active enZYmes database, which include LPMOs from bacteria, fungi, insects, and viruses. LPMOs have become important enzymes both industrially and scientifically and, in this chapter, we provide a brief introduction to chitin-active LPMOs including a summary of the 20+ chitin-active LPMOs that have been characterized so far. Then, we describe their structural features, catalytic mechanism, and appended carbohydrate modules. Finally, we show how chitin-active LPMOs can be used to perform chemo-enzymatic modification of chitin substrates.


Subject(s)
Chitin/chemistry , Mixed Function Oxygenases , Animals , Bacteria/enzymology , Cell Wall , Fungi/enzymology , Glycoside Hydrolases , Insecta/enzymology , Viruses/enzymology
11.
Biochim Biophys Acta Mol Cell Res ; 1864(11 Pt A): 2026-2035, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28392403

ABSTRACT

The matrix metalloproteinase (MMP) family belongs to the metzincin clan of zinc-dependent metallopeptidases. Due to their enormous implications in physiology and disease, MMPs have mainly been studied in vertebrates. They are engaged in extracellular protein processing and degradation, and present extensive paralogy, with 23 forms in humans. One characteristic of MMPs is a ~165-residue catalytic domain (CD), which has been structurally studied for 14 MMPs from human, mouse, rat, pig and the oral-microbiome bacterium Tannerella forsythia. These studies revealed close overall coincidence and characteristic structural features, which distinguish MMPs from other metzincins and give rise to a sequence pattern for their identification. Here, we reviewed the literature available on MMPs outside vertebrates and performed database searches for potential MMP CDs in invertebrates, plants, fungi, viruses, protists, archaea and bacteria. These and previous results revealed that MMPs are widely present in several copies in Eumetazoa and higher plants (Tracheophyta), but have just token presence in eukaryotic algae. A few dozen sequences were found in Ascomycota (within fungi) and in double-stranded DNA viruses infecting invertebrates (within viruses). In contrast, a few hundred sequences were found in archaea and >1000 in bacteria, with several copies for some species. Most of the archaeal and bacterial phyla containing potential MMPs are present in human oral and gut microbiomes. Overall, MMP-like sequences are present across all kingdoms of life, but their asymmetric distribution contradicts the vertical descent model from a eubacterial or archaeal ancestor. This article is part of a Special Issue entitled: Matrix Metalloproteinases edited by Rafael Fridman.


Subject(s)
Archaea/enzymology , Archaeal Proteins , Bacteria/enzymology , Bacterial Proteins , Invertebrates/enzymology , Matrix Metalloproteinases , Viral Proteins , Viruses/enzymology , Animals , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Matrix Metalloproteinases/chemistry , Matrix Metalloproteinases/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism
12.
Article in English | MEDLINE | ID: mdl-29061751

ABSTRACT

A single dose of laninamivir octanoate (LO) inhaled using a dry powder inhaler (DPI) is effective for the treatment and prophylaxis of influenza. Nebulizers are an option for pediatric and elderly patients who may have difficulty in using a DPI. A single-center, open-label study was conducted to evaluate the plasma and intrapulmonary pharmacokinetics (PK) of laninamivir after a single nebulized administration of LO in healthy male Japanese subjects for identifying a safe and effective dosage regimen for a nebulizer. A single dose of LO (40 to 320 mg) was administered using a nebulizer, and plasma concentrations of LO and laninamivir were analyzed up to 168 h after inhalation by validated liquid chromatography-tandem mass spectrometry methods. Subgroups of 6 subjects each underwent bronchoalveolar lavage at specified time intervals over 4 to 168 h following a single nebulized administration of LO (160 mg), and the concentrations in epithelial lining fluid (ELF) were calculated by the urea diffusion method. PK parameters were determined by noncompartment analysis. Inhaled nebulized LO was found to be safe and well tolerated up to the highest dose evaluated (320 mg). Plasma laninamivir concentrations increased almost dose proportionally. Laninamivir concentrations in ELF exceeded the 50% inhibitory concentrations for viral neuraminidase up to 168 h after the nebulized inhalation of 160 mg LO. Thus, similarly to the DPI, ELF concentration profiles of laninamivir after a single nebulized administration support its long-lasting effect against influenza virus infection. This study has been registered at JAPIC Clinical Trials Information (http://www.clinicaltrials.jp/) under registration no. JAPIC CTI-152996.


Subject(s)
Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Neuraminidase/antagonists & inhibitors , Zanamivir/analogs & derivatives , Administration, Inhalation , Adult , Antiviral Agents/adverse effects , Asian People , Bronchoalveolar Lavage Fluid/chemistry , Dose-Response Relationship, Drug , Dry Powder Inhalers , Guanidines , Healthy Volunteers , Humans , Male , Middle Aged , Nebulizers and Vaporizers , Pyrans , Sialic Acids , Viruses/drug effects , Viruses/enzymology , Young Adult , Zanamivir/administration & dosage , Zanamivir/adverse effects , Zanamivir/pharmacokinetics
13.
Biochim Biophys Acta Proteins Proteom ; 1866(1): 141-154, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28502748

ABSTRACT

Sequencing in all areas of the tree of life has produced >300,000 cytochrome P450 (CYP) sequences that have been mined and collected. Nomenclature has been assigned to >41,000 CYP sequences and the majority of the remainder has been sorted by BLAST searches into clans, families and subfamilies in preparation for naming. The P450 sequence space is being systematically explored and filled in. Well-studied groups like vertebrates are covered in greater depth while new insights are being added into uncharted territories like horseshoe crab (Limulus polyphemus), tardigrades (Hypsibius dujardini), velvet worm (Euperipatoides_rowelli), and basal land plants like hornworts, liverworts and mosses. CYPs from the fungi, one of the most diverse groups, are being explored and organized as nearly 800 fungal species are now sequenced. The CYP clan structure in fungi is emerging with 805 CYP families sorting into 32 CYP clans. >3000 bacterial sequences are named, mostly from terrestrial or freshwater sources. Of 18,379 bacterial sequences downloaded from the CYPED database, all are >43% identical to named CYPs. Therefore, they fit in the 602 named P450 prokaryotic families. Diversity in this group is becoming saturated, however 25% of 3305 seawater bacterial P450s did not match known P450 families, indicating marine bacterial CYPs are not as well sampled as land/freshwater based bacterial CYPs. Future sequencing plans of the Genome 10K project, i5k and GIGA (Global Invertebrate Genomics Alliance) are expected to produce more than one million cytochrome P450 sequences by 2020. This article is part of a Special Issue entitled: Cytochrome P450 biodiversity and biotechnology, edited by Erika Plettner, Gianfranco Gilardi, Luet Wong, Vlada Urlacher, Jared Goldstone.


Subject(s)
Cytochrome P-450 Enzyme System/classification , Cytochrome P-450 Enzyme System/genetics , Genetic Variation , Genome , Phylogeny , Animals , Archaea/classification , Archaea/enzymology , Archaea/genetics , Arthropods/classification , Arthropods/enzymology , Arthropods/genetics , Bacteria/classification , Bacteria/enzymology , Bacteria/genetics , Biological Evolution , Birds/classification , Birds/genetics , Birds/metabolism , Cytochrome P-450 Enzyme System/metabolism , Fungi/classification , Fungi/enzymology , Fungi/genetics , Gene Expression , Humans , Multigene Family , Plants/classification , Plants/enzymology , Plants/genetics , Tardigrada/classification , Tardigrada/enzymology , Tardigrada/genetics , Viruses/classification , Viruses/enzymology , Viruses/genetics
14.
Crit Rev Microbiol ; 44(3): 304-317, 2018 May.
Article in English | MEDLINE | ID: mdl-28885059

ABSTRACT

Host anti-viral innate-immune signalling pathways are regulated by a variety of post-translation modifications including ubiquitination, which is critical to regulate various signalling pathways for synthesis of anti-viral molecules. A homeostasis of host immune responses, induced due to viral infection and further ubiquitination, is maintained by the action of deubiquitinases (DUB). Infecting viruses utilize the process of deubiquitination for tricking host immune system wherein viral DUBs compete with host DUBs for inhibition of innate-immune anti-viral signalling pathways, which instead of maintaining an immune homeostasis bring about virus-mediated pathogenesis. This suggests that viruses co-evolve with their hosts to acquire similar machinery for tricking immune surveillance and establishing infection.


Subject(s)
Deubiquitinating Enzymes/immunology , Viral Proteins/immunology , Virus Diseases/immunology , Viruses/enzymology , Animals , Deubiquitinating Enzymes/genetics , Host-Pathogen Interactions , Humans , Immune Evasion , Immunity, Innate , Viral Proteins/genetics , Virus Diseases/enzymology , Virus Diseases/genetics , Virus Diseases/virology , Viruses/genetics , Viruses/immunology
17.
RNA Biol ; 14(10): 1314-1319, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28277928

ABSTRACT

The nucleotide addition cycle of nucleic acid polymerases includes 2 major events: the pre-chemistry active site closure leading to the addition of one nucleotide to the product chain; the post-chemistry translocation step moving the polymerase active site one position downstream on its template. In viral RNA-dependent RNA polymerases (RdRPs), structural and biochemical evidences suggest that these 2 events are not tightly coupled, unlike the situation observed in A-family polymerases such as the bacteriophage T7 RNA polymerase. Recently, an RdRP translocation intermediate crystal structure of enterovirus 71 shed light on how translocation may be controlled by elements within RdRP catalytic motifs, and a series of poliovirus apo RdRP crystal structures explicitly suggest that a motif B loop may assist the movement of the template strand in late stages of transcription. Implications of RdRP catalysis-translocation uncoupling and the remaining challenges to further elucidate RdRP translocation mechanism are also discussed.


Subject(s)
RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Viruses/enzymology , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Protein Conformation , Protein Transport , Viral Proteins/chemistry , Viral Proteins/metabolism , Viruses/chemistry
18.
Nucleic Acids Res ; 43(14): 6651-64, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26109351

ABSTRACT

Until relatively recently, DNA primases were viewed simply as a class of proteins that synthesize short RNA primers requisite for the initiation of DNA replication. However, recent studies have shown that this perception of the limited activities associated with these diverse enzymes can no longer be justified. Numerous examples can now be cited demonstrating how the term 'DNA primase' only describes a very narrow subset of these nucleotidyltransferases, with the vast majority fulfilling multifunctional roles from DNA replication to damage tolerance and repair. This article focuses on the archaeo-eukaryotic primase (AEP) superfamily, drawing on recently characterized examples from all domains of life to highlight the functionally diverse pathways in which these enzymes are employed. The broad origins, functionalities and enzymatic capabilities of AEPs emphasizes their previous functional misannotation and supports the necessity for a reclassification of these enzymes under a category called primase-polymerases within the wider functional grouping of polymerases. Importantly, the repositioning of AEPs in this way better recognizes their broader roles in DNA metabolism and encourages the discovery of additional functions for these enzymes, aside from those highlighted here.


Subject(s)
DNA Primase/metabolism , DNA Repair Enzymes/metabolism , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Archaea/enzymology , DNA Damage , DNA Primase/chemistry , DNA Primase/classification , DNA Primase/genetics , DNA Repair , DNA Repair Enzymes/chemistry , DNA-Directed DNA Polymerase/chemistry , Eukaryota/enzymology , Evolution, Molecular , Humans , Plasmids/biosynthesis , Trypanosoma/enzymology , Viruses/enzymology
19.
Adv Exp Med Biol ; 925: 75-87, 2017.
Article in English | MEDLINE | ID: mdl-27677277

ABSTRACT

Glycosaminoglycans (GAGs) are important constituents of the extracellular matrix that make significant contributions to biological processes and have been implicated in a wide variety of diseases. GAG-degrading enzymes with different activities have been found in various animals and microorganisms, and they play an irreplaceable role in the structure and function studies of GAGs. As two kind of important GAG-degrading enzymes, hyaluronidase (HAase) and chondroitinase (CSase) have been widely studied and increasing evidence has shown that, in most cases, their substrate specificities overlap and thus the "HAase" or "CSase" terms may be improper or even misnomers. Different from previous reviews, this article combines HAase and CSase together to discuss the traditional classification, substrate specificity, degradation pattern, new resources and naming of these enzymes.


Subject(s)
Chondroitinases and Chondroitin Lyases/chemistry , Eukaryotic Cells/chemistry , Extracellular Matrix/chemistry , Glycosaminoglycans/metabolism , Hyaluronoglucosaminidase/chemistry , Animals , Bacteria/chemistry , Bacteria/enzymology , Carbohydrate Conformation , Carbohydrate Sequence , Chondroitinases and Chondroitin Lyases/classification , Chondroitinases and Chondroitin Lyases/isolation & purification , Chondroitinases and Chondroitin Lyases/metabolism , Eukaryotic Cells/cytology , Glycosaminoglycans/chemistry , Humans , Hyaluronoglucosaminidase/classification , Hyaluronoglucosaminidase/isolation & purification , Hyaluronoglucosaminidase/metabolism , Hydrolysis , Kinetics , Substrate Specificity , Viruses/chemistry , Viruses/enzymology
20.
Proc Jpn Acad Ser B Phys Biol Sci ; 93(7): 449-463, 2017.
Article in English | MEDLINE | ID: mdl-28769016

ABSTRACT

Favipiravir (T-705; 6-fluoro-3-hydroxy-2-pyrazinecarboxamide) is an anti-viral agent that selectively and potently inhibits the RNA-dependent RNA polymerase (RdRp) of RNA viruses. Favipiravir was discovered through screening chemical library for anti-viral activity against the influenza virus by Toyama Chemical Co., Ltd. Favipiravir undergoes an intracellular phosphoribosylation to be an active form, favipiravir-RTP (favipiravir ribofuranosyl-5'-triphosphate), which is recognized as a substrate by RdRp, and inhibits the RNA polymerase activity. Since the catalytic domain of RdRp is conserved among various types of RNA viruses, this mechanism of action underpins a broader spectrum of anti-viral activities of favipiravir. Favipiravir is effective against a wide range of types and subtypes of influenza viruses, including strains resistant to existing anti-influenza drugs. Of note is that favipiravir shows anti-viral activities against other RNA viruses such as arenaviruses, bunyaviruses and filoviruses, all of which are known to cause fatal hemorrhagic fever. These unique anti-viral profiles will make favipiravir a potentially promising drug for specifically untreatable RNA viral infections.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , DNA-Directed RNA Polymerases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Pyrazines/pharmacology , Viruses/enzymology , Animals , Humans , Viruses/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL