Leta i den här bloggen


fredag 29 maj 2020

Tuman Lamiinit

Naetar N, Ferraioli S, Foisner R. 2017. Lamins in the nuclear interior - life outside the lamina. J. Cell Sci. 130(13): 2087-2096 Crossref, Medline, ISIGoogle Scholar.

onsdag 27 maj 2020

ZGRF, GRF-tyyppiset sinkkisormiproteiinit ovat seitsemän ZGRF.geenin koodaamia

ZGRF1, (4q25). Zinc  Finger GRF-type containing.  Nucleic acid binding domain:  in the middle there are  three   conserved aminoacids G, R and F as a motif in this group of Zinc finger proteins.
 https://www.genecards.org/cgi-bin/carddisp.pl?gene=ZGRF1&keywords=ZGRF1


ZGFR2,(Xp11.2),  APEX2 , DNA-(apurinic or apyrimidinic site) lyase 2 , AP endonuclease. https://www.genecards.org/cgi-bin/carddisp.pl?gene=APEX2&keywords=ZGRF2
 Function as a weak apurinic/apyrimidinic (AP) endodeoxyribonuclease in the DNA base excision repair (BER) pathway of DNA lesions induced by oxidative and alkylating agents. Initiates repair of AP sites in DNA by catalyzing hydrolytic incision of the phosphodiester backbone immediately adjacent to the damage, generating a single-strand break with 5'-deoxyribose phosphate and 3'-hydroxyl ends. Displays also double-stranded DNA 3'-5' exonuclease, 3'-phosphodiesterase activities. Shows robust 3'-5' exonuclease activity on 3'-recessed heteroduplex DNA and is able to remove mismatched nucleotides preferentially. Shows fairly strong 3'-phosphodiesterase activity involved in the removal of 3'-damaged termini formed in DNA by oxidative agents. In the nucleus functions in the PCNA-dependent BER pathway. Required for somatic hypermutation (SHM) and DNA cleavage step of class switch recombination (CSR) of immunoglobulin genes. Required for proper cell cycle progression during proliferation of peripheral lymphocytes.
APEX2_HUMAN,Q9UBZ4
ZGFR3, (4q34.3),  Endonuclease 8 like 3.  NEIL3.
 NEIL3i like DNA glycosylase 3 (initiating ( The  first step in BER Base Excision  Repair) ( Contains even  Zf-RanBP2-like domain as  ZRANB )
 https://www.genecards.org/cgi-bin/carddisp.pl?gene=NEIL3&keywords=ZGRF3
 DNA glycosylase which prefers single-stranded DNA (ssDNA), or partially ssDNA structures such as bubble and fork structures, to double-stranded DNA (dsDNA). In vitro, displays strong glycosylase activity towards the hydantoin lesions spiroiminodihydantoin (Sp) and guanidinohydantoin (Gh) in both ssDNA and dsDNA; also recognizes FapyA, FapyG, 5-OHU, 5-OHC, 5-OHMH, Tg and 8-oxoA lesions in ssDNA. No activity on 8-oxoG detected. Also shows weak DNA-(apurinic or apyrimidinic site) lyase activity. In vivo, appears to be the primary enzyme involved in removing Sp and Gh from ssDNA in neonatal tissues. Seems to be an important facilitator of cell proliferation in certain populations, for example neural stem/progenitor cells and tumor cells, suggesting a role in replication-associated DNA repair. NEIL3_HUMAN,Q8TAT5

ZGFR4, (4p15.2), Recommended name: rRNA N6-adenosine-methyltransferase ZCCHC4
ZCCHC4, Even  DHHC -domain containig  Znf protein (conserved domain  409- 438 Zf- DHHC palmityltransferase)
  https://www.genecards.org/cgi-bin/carddisp.pl?gene=ZCCHC4&keywords=ZGRF4

ZGFR5, (16p12.3), ERI2,   Recommended name: ERI1 exoribonuclease 2
https://www.genecards.org/cgi-bin/carddisp.pl?gene=ERI2&keywords=ZGRF5

ZGFR6, (1p13.1), TTF2,  Recommended name:Transcription termination factor 2
 https://www.genecards.org/cgi-bin/carddisp.pl?gene=TTF2&keywords=ZGRF6   This gene encodes a member of the SWI2/SNF2 family of proteins, which play a critical role in altering protein-DNA interactions. The encoded protein has been shown to have dsDNA-dependent ATPase activity and RNA polymerase II termination activity. This protein interacts with cell division cycle 5-like, associates with human splicing complexes, and plays a role in pre-mRNA splicing. [provided by RefSeq, Jul 2008]  Aldosteronisynthesis  pathways.

ZGFR7, (17p11.2), TOP3A, DNA topoisomerase  III alpha. (Contains  ssDNA binding  Zn finger and  Zn ribbon)   Recommended name:  DNA topoisomerase 3-alpha
https://www.genecards.org/cgi-bin/carddisp.pl?gene=TOP3A&keywords=ZGRF7

måndag 25 maj 2020

SMAD perheestä Esimerkkejä TGFbeta /SMAD säätelystä.

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5104937/

SMAD  fylogeneettinen kaava:
Generation of GFP-SMAD cell lines. A, SMAD protein phylogeny. Clustering analysis and systemic representation of SMAD proteins with the indicated domains (MH1, MH2, and SAD). SMAD categorization is indicated on the right.
 B, SMAD cell line time series after doxycycline induction was analyzed on immunoblot. HeLa cell lines were induced with 1 μg/ml of doxycycline for 4, 8, 16, 20, or 24 h prior to harvesting or harvested without induction. Blots were probed using GFP or GAPDH antibodies.
 An external file that holds a picture, illustration, etc.
Object name is zbc0501656480001.jpg

Discussion

SMAD signaling is involved in many different cellular processes. In this study interactome datasets for all SMAD proteins were generated using affinity purification for HeLa cells followed by mass spectrometry analysis. The nuclear transport protein IPO5 was identified as a novel interactor for BMP-regulated SMAD1 and using confocal microscopy a functional link was shown between IPO5 and the BMP-regulated R-SMADs but not TGF-β-regulated R-SMADs. By extending the NLS lysine stretch in SMAD3 we found SMAD3 nuclear localization sensitive to IPO5, suggesting that the length of the lysine stretch in the NLS is responsible for differentiation between the BMP- and TGF-β-regulated SMADs.

SMAD proteins shuttle continuously between the cytoplasm and nucleus, which is independent of a receptor activation signal ().  

The export of SMAD4 is known to be CRM1-mediated, whereas export of R-SMADs is CRM1-independent.https://pubmed.ncbi.nlm.nih.gov/28399435/
XPO1 (2p15), CRM1, Exportin 1

 Phosphorylation of R-SMAD followed by SMAD4 recruitment causes nuclear accumulation of SMADs, which result in reduced nuclear export.

 Transcription factors like TAZ, Tafazzin (Xq28)  () have been shown to be responsible for SMAD nuclear accumulation, although the exact mechanism is unknown.
Conserv. domain: Lysophospholipid Acyltransferases (LPLATs) of Glycerophospholipid Biosynthesis: AGPAT-like
Lysophospholipid acyltransferase (LPLAT) superfamily member: acyltransferases of de novo and remodeling pathways of glycerophospholipid biosynthesis which catalyze the incorporation of an acyl group from either acylCoAs or acyl-acyl carrier proteins (acylACPs) into acceptors such as glycerol 3-phosphate, dihydroxyacetone phosphate or lyso-phosphatidic acid. Included in this subgroup are such LPLATs as 1-acyl-sn-glycerol-3-phosphate acyltransferase (AGPAT, PlsC), Tafazzin (product of Barth syndrome gene), and similar proteins.
  TITLE     Defective Mitochondrial Cardiolipin Remodeling Dampens HIF-1alpha
            Expression in Hypoxia
  JOURNAL   Cell Rep 25 (3), 561-570 (2018)
   PUBMED   30332638
  REMARK    GeneRIF: Tafazzin deficiency is associated with defective
            remodeling of the mitochondrial phospholipid cardiolipin causing
            cardiomyopathy in Barth syndrome.

 In the nucleus, phosphorylation of R-SMAD complexes is gradually reduced causing dissociation of SMAD4 followed by export from the nucleus. This constant SMAD nuclear/cytoplasmic cycling is an important control mechanism for constant monitoring of the receptor activation status. This results in a quick adaptation of nuclear SMAD accumulation, when the signal is present and to reduce to steady state levels when the receptors become inactive.

 Initially, transport proteins importin α1* (KBNA1)  and β1* (KBNB1)  () have been connected with SMAD signaling. 

Later studies implied IPO7** and IPO8** in signal-dependent and -independent transport of SMAD1/3/4 involving NLS found in the MH1 domain.

 Our results corroborate an important role of IPO7 in subcellular distribution of SMAD1, -2, -3, -5, and -9 (Fig. 7). IPO7 seems to be the major contributor of SMAD nuclear import, whereas the import mediated by IPO5 is restricted. Nuclear import of the R-SMADs by IPO7 does not select for a specific type of R-SMAD (Fig. 7). Systemically testing IPO5 against all SMADs in this study shows selectivity of IPO5 for BMP-activated R-SMADs, which is determined by the length of the lysine stretch of the NLS. Selectivity of the IPO5 protein for the cargo allows for regulation of signaling, thus adding a new player in controlling the subcellular distribution of SMAD proteins.

TÄSSÄ sivuhyppy: Haen eri  IPO proteiineja ja  importiini alfa 1 ja importiini alfa 2 ja importiini beta proteiineja esiin. Merkkaan  tähdeää ne joita  on tarkoitettu tekstissä:

 //      IPO1*, IPOB, (17q21.32)   Importin subunit B1*,    Karyopherin subunit beta 1.
UniProtKB/Swiss-Prot Summary for KPNB1 Gene:
  • Functions in nuclear protein import, either in association with an adapter protein, like an importin-alpha subunit, which binds to nuclear localization signals (NLS) in cargo substrates, or by acting as autonomous nuclear transport receptor. Acting autonomously, serves itself as NLS receptor. Docking of the importin/substrate complex to the nuclear pore complex (NPC) is mediated by KPNB1 through binding to nucleoporin FxFG repeats and the complex is subsequently translocated through the pore by an energy requiring, Ran-dependent mechanism. At the nucleoplasmic side of the NPC, Ran binds to importin-beta and the three components separate and importin-alpha and -beta are re-exported from the nucleus to the cytoplasm where GTP hydrolysis releases Ran from importin. The directionality of nuclear import is thought to be conferred by an asymmetric distribution of the GTP- and GDP-bound forms of Ran between the cytoplasm and nucleus. Mediates autonomously the nuclear import of ribosomal proteins RPL23A, RPS7 and RPL5. Binds to a beta-like import receptor binding (BIB) domain of RPL23A. In association with IPO7 mediates the nuclear import of H1 histone. In vitro, mediates nuclear import of H2A, H2B, H3 and H4 histones. In case of HIV-1 infection, binds and mediates the nuclear import of HIV-1 Rev. Imports SNAI1 and PRKCI into the nucleus.
 IPO2, TNPO1, Transportin 1, KPNB2 (5q13.2) MIP1, MIP, TRN.
 
IPO3, TNPO2, (19p13.13). Transportin 2. Karyopherin beta 2b, (KPNB2b) 

IPO4, Importin 4b , (14q12), RANBP4, IMP4B. , Ran -binding protein 4. https://www.genecards.org/cgi-bin/carddisp.pl?gene=IPO4&keywords=IPO4

IPO5* , Importin 5, Karyopherin beta 3, (KPNB3), RANBP5.   https://www.genecards.org/cgi-bin/carddisp.pl?gene=IPO5&keywords=IPO5
 
IPO9 

IPOA1,Pendulin,Karyopherin alpha 2,(KPNA2),(17q24.2),  SRP1alpha, RAG cohort protein 1.   

IPOA2

IPOA3, Importin subunit alpha 3,( KPNA4), Qip1, (3q25.33), Importin subunit alpha 3. 
IPOA4, ( KPNA3),  Qip2, (13q14.2).Importin subunit alpha 4. 

IPOA5: (3q21.1) ( KPNA1), Karyopherin subunit alpha 1, Importin subunit alpha 5. RAG cohort protein 2, recombination activating gene cohort 2. 
The transport of molecules between the nucleus and the cytoplasm in eukaryotic cells is mediated by the nuclear pore complex (NPC), which consists of 60-100 proteins. Small molecules (up to 70 kD) can pass through the nuclear pore by nonselective diffusion while larger molecules are transported by an active process. The protein encoded by this gene belongs to the importin alpha family, and is involved in nuclear protein import. This protein interacts with the recombination activating gene 1 (RAG1) protein and is a putative substrate of the RAG1 ubiquitin ligase. Alternative splicing results in multiple transcript variants. [provided by RefSeq, Nov 2012] 
( Näitä näyttää olevan runsaasti Gene Cards lähteessä ja monen yhteydssä mainitaan viruksista) -//


The SMAD activation pathway is a signaling cascade that gains complexity in regulation over different levels in the cell (, ,). 

Over 30 different molecules including TGF-β and BMP can be found in the human body that belong to the TGF-β superfamily (). These molecules can signal to a combination of seven type I and five type II receptors. Because not all of these receptors are present in each cell type this makes TGF-β and BMP signaling very context-dependent. It is important to note that we did not identify any TGF-β or BMP receptor in GFP-SMAD purifications from the cytoplasmic fractions, which could be due to the low efficiency of membrane protein extraction in our procedures.
The N-terminal location of the GFP moiety in the SMAD fusion constructs was chosen to maintain the C-terminal SSXS residues required for R-SMAD phosphorylation and protein activation (,). This resulted in fusion constructs that could be phosphorylated upon stimulation of their receptors using their respective ligands (Fig. 3). However, the possibility still remains that interaction partners have been missed due to N-terminal tagging of SMAD proteins.


In our experiments we confirmed binding of the transcriptional repressor LEMD3 to SMAD3 in the nucleus and provide evidence that LEMD3 and SMAD4 binding are mutually exclusive (Fig. 5). 
 The nuclear envelope LEM-domain protein emerin
LEMD3 (12q14.3), Inner nuclear membrane protein Man1.tekee interaktion SMAD1,2,3 ja 5 kanssa ja sitoutuu sekä fosforyloituun että fosforyloitumattomaan R-SMADiin.
 Transforming Growth Factor Beta and Bone: Lessons Learned from ...

 Interestingly, no LEMD3 binding to other R-SMADs is observed, whereas SKI and SKIL are identified in SMAD2/3 and -4, which indicates a high specificity in repressor interactions.


Activated SMAD proteins stimulate gene transcription, but the exact nature of their nuclear coactivators has not been determined in much detail. We provide evidence that Mediator, TFIID, and SET/MLL but not BAF complex could participate in transcription activation by the SMAD1 and SMAD2 proteins. The observation of specific co-activators for specific SMAD family members warrants a further study into the downstream transcription activation pathways for TGF-β and BMP signaling.


( Alla oleva artikkeli selvittää SMAD3:n osuuta  mikroRNA-säätelyssä, esimerkki on munuaisfibroosin terapiasta: 
Article in Frontiers in Physiology · March 2015
DOI: 10.3389/fphys.2015.00082 ·)

söndag 24 maj 2020

TRPS1 ((8q23.3), LGCR, GC79 , ( GATAD- ryhmän sinkkisormiproteiineista )

 GATAD -ryhmän inkkisormiproteiinit ovat  luvultaan 15 ja niistä kaikki 15  ovat transkriptiotekijöitä.  GATAD-ryhmän sinkkisormiproteiinit  sitoutuvat  DNA-konsensusdomeeniin  (AT)GATA(AG). Tämä proteiini toimii  transkriptionaalisena repressorina   ja myös kiinnittyy dyneiiniproteiinin kevyeeseen ketjuun. Dyneiinillä on  GATA välitteinen säätely.
 SARS-2 viruksen   oletettu mikroRNA  pitää tätä proteiinia TRPS1 kohdeproteiinina.
  895..938
                     /region_name="ZnF_GATA"
                     /note="zinc finger binding to DNA consensus sequence
                     [AT]GATA[AG]; smart00401"
                     /db_xref="CDD:214648"


https://www.ncbi.nlm.nih.gov/protein/NP_001269831.1
   Genomics 29 (1), 87-97 (1995)
   PUBMED   8530105
REFERENCE   9  (residues 1 to 1285)
  AUTHORS   Maas,S., Shaw,A., Bikker,H. and Hennekam,R.C.M.
  TITLE     Trichorhinophalangeal Syndrome
  JOURNAL   (in) Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Stephens
            K and Amemiya A (Eds.);
            GENEREVIEWS((R));
            (1993)
   PUBMED   28426188
REFERENCE   10 (residues 1 to 1285)
  AUTHORS   Hamers A, Jongbloet P, Peeters G, Fryns JP and Geraedts J.
  TITLE     Severe mental retardation in a patient with tricho-rhino-phalangeal
            syndrome type I and 8q deletion
  JOURNAL   Eur. J. Pediatr. 149 (9), 618-620 (1990)
   PUBMED   2373110
COMMENT     REVIEWED REFSEQ: This record has been curated by NCBI staff. The
            reference sequence was derived from AF178030.3, DC339831.1,
            AK304046.1, AF183810.1, KF454923.1 and KC877306.1.
            On Sep 21, 2013 this sequence version replaced XP_005251105.1.
            
            Summary: This gene encodes a transcription factor that represses
            GATA-regulated genes and binds to a dynein light chain protein.
            Binding of the encoded protein to the dynein light chain protein
            affects binding to GATA consensus sequences and suppresses its
            transcriptional activity. Defects in this gene are a cause of
            tricho-rhino-phalangeal syndrome (TRPS) types I-III. [provided by
            RefSeq, Jul 2008].
            
            Transcript Variant: This variant (2) uses an alternate 5' structure
            which results in the use of an alternate start codon, compared to
            variant 1. The encoded isoform (2) is shorter and has a distinct
            N-terminus, compared to isoform 1.
            
            Sequence Note: This RefSeq record was created from transcript and
            genomic sequence data to make the sequence consistent with the
            reference genome assembly. The genomic coordinates used for the
            transcript record were based on transcript alignments.
            
            Publication Note:  This RefSeq record includes a subset of the
            publications that are available for this gene. Please see the Gene
            record to access additional publications.
            
            ##Evidence-Data-START##
            Transcript exon combination :: SRR1803614.6752.1, AK304046.1
                                           [ECO:0000332]
            RNAseq introns              :: single sample supports all introns
                                           SAMEA2467148, SAMEA2467150
                                           [ECO:0000348]
            ##Evidence-Data-END##
FEATURES             Location/Qualifiers
     source          1..1285
                     /organism="Homo sapiens"
                     /db_xref="taxon:9606"
                     /chromosome="8"
                     /map="8q23.3"
     Protein         1..1285
                     /product="zinc finger transcription factor Trps1 isoform
                     2"
                     /note="zinc finger transcription factor Trps1; zinc finger
                     protein GC79; trichorhinophalangeal syndrome I;
                     tricho-rhino-phalangeal syndrome type I protein"
                     /calculated_mol_wt=141851
     Region          895..938
                     /region_name="ZnF_GATA"
                     /note="zinc finger binding to DNA consensus sequence
                     [AT]GATA[AG]; smart00401"
                     /db_xref="CDD:214648"
     Region          899..955
                     /region_name="ZnF_GATA"
                     /note="Zinc finger DNA binding domain; binds specifically
                     to DNA consensus sequence [AT]GATA[AG] promoter elements;
                     a subset of family members may also bind protein;
                     zinc-finger consensus topology is C-X(2)-C-X(17)-C-X(2)-C;
                     cd00202"
                     /db_xref="CDD:238123"
     Site            order(900,903,921,924)
                     /site_type="other"
                     /note="zinc binding site [ion binding]"
                     /db_xref="CDD:238123"
     Site            order(909..910,912,921..931,952,954..955)
                     /site_type="DNA binding"
                     /note="DNA-binding region [nucleotide binding]"
                     /db_xref="CDD:238123"
     CDS             1..1285
                     /gene="TRPS1"
                     /gene_synonym="GC79; LGCR"
                     /coded_by="NM_001282902.3:131..3988"
                     /note="isoform 2 is encoded by transcript variant 2"
                     /db_xref="CCDS:CCDS64957.1"
                     /db_xref="GeneID:7227"
                     /db_xref="HGNC:HGNC:12340"
                     /db_xref="MIM:604386"

torsdag 21 maj 2020

Antiviraalista MxA:sta asiaa . Haku " MxA, coronavirus" ( 11 vastausta)


Search Results
11 results


Middle East respiratory syndrome coronavirus shows poor replication but significant induction of antiviral responses in human monocyte-derived macrophages and dendritic cells.
Tynell J, Westenius V, Rönkkö E, Munster VJ, Melén K, Österlund P, Julkunen I. J Gen Virol. 2016 Feb;97(2):344-355. doi: 10.1099/jgv.0.000351. Epub 2015 Nov 24. PMID: 26602089 Free PMC article.
n this study we assessed the ability of Middle East respiratory syndrome coronavirus (MERS-CoV) to replicate and induce innate immunity in human monocyte-derived macrophages and dendritic cells (MDDCs), and compared it with severe acute respiratory syndrome coronavirus (SARS-CoV). Assessments of viral protein and RNA levels in infected cells showed that both viruses were impaired in their ability to replicate in these cells. Some induction of IFN-λ1, CXCL10 and MxA mRNAs in both macrophages and MDDCs was seen in response to MERS-CoV infection, but almost no such induction was observed in response to SARS-CoV infection. ELISA and Western blot assays showed clear production of CXCL10 and MxA in MERS-CoV-infected macrophages and MDDCs. Our data suggest that SARS-CoV and MERS-CoV replicate poorly in human macrophages and MDDCs, but MERS-CoV is nonetheless capable of inducing a readily detectable host innate immune response. Our results highlight a clear difference between the viruses in activating host innate immune responses in macrophages and MDDCs, which may contribute to the pathogenesis of infection. 
 
2. 
IFN-lambda preferably inhibits PEDV infection of porcine intestinal epithelial cells compared with IFN-alpha.
Li L, Fu F, Xue M, Chen W, Liu J, Shi H, Chen J, Bu Z, Feng L, Liu P. Antiviral Res. 2017 Apr;140:76-82. doi: 10.1016/j.antiviral.2017.01.012. Epub 2017 Jan 19. PMID: 28109912 Free PMC article.
In contrast to type I interferons that target various types of cells and organs, interferon lambda (IFN-L) primarily acts on mucosal epithelial cells and exhibits robust antiviral activity within the mucosal surface. Porcine epidemic diarrhea virus (PEDV), which causes high morbidity and mortality in piglets, is an enteropathogenic coronavirus with economic importance. Here, we demonstrated that both recombinant porcine IFN-L1 (rpIFN-L1) and rpIFN-L3 have powerful antiviral activity against PEDV infection of both Vero E6 cells and the intestinal porcine epithelial cell line J2 (IPEC-J2). Both forms of rpIFN-L inhibited two genotypes of PEDV (strain CV777 of genotype 1 and strain LNCT2 of genotype 2). rpIFN-L1 primarily controlled viral infection in the early stage and had less antiviral activity in IPEC-J2 than in rpIFN-L3 cells infected with PEDV. In addition, rpIFN-L1 exhibited greater antiviral activity against PEDV infection of IPEC-J2 cells than that of porcine IFN-alpha. Consistent with this finding, rpIFN-L1 triggered higher levels of certain antiviral IFN-stimulated genes (ISGs) (ISG15, OASL, and MxA) in IPEC-J2 cells than porcine IFN-alpha. Although IPEC-J2 cells responded to both IFN-alpha and lambda, transcriptional profiling of ISGs (specifically ISG15, OASL, MxA, and IFITMs) differed when induced by either IFN-alpha or rpIFN-L. Therefore, our data provide the experimental evidence that porcine IFN-L suppresses PEDV infection of IPEC-J2 cells, which may offer a promising therapeutic for combating PED in piglets. 
 
3.
Blood MxA protein as a marker for respiratory virus infections in young children.
Toivonen L, Schuez-Havupalo L, Rulli M, Ilonen J, Pelkonen J, Melen K, Julkunen I, Peltola V, Waris M. J Clin Virol. 2015 Jan;62:8-13. doi: 10.1016/j.jcv.2014.11.018. Epub 2014 Nov 18. PMID: 25542463 Free PMC article.
BACKGROUND: Type I interferon induced MxA response can differentiate viral from bacterial infections, but MxA responses in rhinovirus or asymptomatic virus infections are not known.
Background: Type I interferon induced MxA response can differentiate viral from bacterial infections, but MxA responses in rhinovirus or asymptomatic virus infections are not known.
Objective: To study MxA protein levels in healthy state and during respiratory virus infection of young children in an observational prospective cohort.
Study design: Blood samples and nasal swabs were collected from 153 and 77 children with and without symptoms of respiratory infections, respectively. Blood MxA protein levels were measured by an enzyme immunoassay and PCR methods were used for the detection of respiratory viruses in nasal swabs.
Results: Respiratory viruses were detected in 81% of symptomatic children. They had higher blood MxA protein levels (median [interquartile range]) than asymptomatic virus-negative children (695 [345-1370] μg/L vs. 110 [55-170] μg/L; p < 0.001). Within asymptomatic children, no significant difference was observed in MxA responses between virus-positive and virus-negative groups. A cut-off level of 175 μg/L had 92% sensitivity and 77% specificity for a symptomatic respiratory virus infection. Rhinovirus, respiratory syncytial virus, parainfluenza virus, influenza virus, coronavirus, and human metapneumovirus infections were associated with elevated MxA responses. Asymptomatic virus-negative children vaccinated with a live virus vaccine had elevated MxA protein levels (240 [120-540] μg/L), but significantly lower than children with an acute respiratory infection, who had not received vaccinations (740 [350-1425] μg/L; p<0 .001="" p=""> Conclusion: Blood MxA protein levels are increased in young children with symptomatic respiratory virus infections, including rhinovirus infections. MxA is an informative general marker for the most common acute virus infections.
Keywords: Interferon; MxA protein; Respiratory virus infection; Rhinovirus; Rotavirus vaccine; STEPS study.

4.
Human β-defensin 2 plays a regulatory role in innate antiviral immunity and is capable of potentiating the induction of antigen-specific immunity.
Kim J, Yang YL, Jang SH, Jang YS. Virol J. 2018 Aug 8;15(1):124. doi: 10.1186/s12985-018-1035-2. PMID: 30089512 Free PMC article.
Background: Antimicrobial peptides (AMPs) are primarily known for their innate immune defense against invading microorganisms, including viruses. In addition, recent research has suggested their modulatory activity in immune induction. Given that most subunit vaccines require an adjuvant to achieve effective immune induction through the activation of innate immunity, AMPs are plausible candidate molecules for stimulating not only innate immune but also adaptive immune responses.
Results: In this study, we investigated the ability of human β-defensin (HBD) 2 to promote antiviral immunity in vitro and in vivo using a receptor-binding domain (RBD) of Middle East respiratory syndrome-coronavirus (MERS-CoV) spike protein (S RBD) as a model antigen (Ag). When HBD 2-conjugated S RBD was used to treat THP-1 human monocytic cells, the expression levels of antiviral (IFN-β, IFN-γ, MxA, PKR, and RNaseL) and primary immune-inducing (NOD2, TNF-α, IL-1β, and IL-6) molecules were enhanced compared to those expressed after treatment with S RBD only. The expression of chemokines capable of recruiting leukocytes, including monocytes/macrophages, natural killer cells, granulocytes, T cells, and dendritic cells, was also increased following HBD 2-conjugated S RBD treatment. More important, immunization of mice with HBD 2-conjugated S RBD enhanced the immunogenicity of the S RBD and elicited a higher S RBD-specific neutralizing antibody response than S RBD alone.
Conclusions: We conclude that HBD 2 activates the primary antiviral innate immune response and may also mediate the induction of an effective adaptive immune response against a conjugated Ag.
Keywords: Adjuvant; Antibody; Antigen; Human β-defensin; MERS-CoV.

5. 
The antiviral effect of interferon-beta against SARS-coronavirus is not mediated by MxA protein.
Spiegel M, Pichlmair A, Mühlberger E, Haller O, Weber F. J Clin Virol. 2004 Jul;30(3):211-3. doi: 10.1016/j.jcv.2003.11.013. PMID: 15135736 Free PMC article.
Severe acute respiratory syndrome (SARS) is caused by a novel coronavirus termed SARS-CoV. No antiviral treatment has been established so far. Interferons are cytokines which induce the synthesis of several antivirally active proteins in the cell. In this study, we demonstrated that multiplication of SARS-CoV in cell culture can be strongly inhibited by pretreatment with interferon-beta. Interferon-alpha and interferon-gamma, by contrast, were less effective. The human MxA protein is one of the most prominent proteins induced by interferon-beta. Nevertheless, no interference with SARS-CoV replication was observed in Vero cells stably expressing MxA. Therefore, other interferon-induced proteins must be responsible for the strong inhibitory effect of interferon-beta against SARS-CoV.
 
 
6. 
Significance of the myxovirus resistance A (MxA) gene -123C>a single-nucleotide polymorphism in suppressed interferon beta induction of severe acute respiratory syndrome coronavirus infection.
Ching JC, Chan KY, Lee EH, Xu MS, Ting CK, So TM, Sham PC, Leung GM, Peiris JS, Khoo US. J Infect Dis. 2010 Jun 15;201(12):1899-908. doi: 10.1086/652799. PMID: 20462354 Free PMC article.
Myxovirus resistance A (MxA) is an antiviral protein induced by interferon alpha and beta (IFN-alpha, IFN-beta) that can inhibit viral replication. The minor alleles of the -88G>T and -123C>A MxA promoter single-nucleotide polymorphisms (SNPs) are associated with increased promoter activity and altered response to IFN-alpha and IFN-beta treatment. Here, we demonstrate that the -123A minor allele provided stronger binding affinity to nuclear proteins extracted from IFN-beta-untreated cells than did the wild-type allele, whereas the -88T allele showed preferential binding after IFN-beta stimulation. Endogenous IFN-alpha and IFN-beta induction can be suppressed in severe acute respiratory syndrome (SARS) coronavirus infection. In support of our in vitro findings, a large case-control genetic-association study for SARS coronavirus infection confirmed that the -123A minor-allele carriers were significantly associated with lower risk of SARS coronavirus infection, whereas the -88T minor-allele carriers were insignificant after adjustment for confounding effects. This suggests that -123C>A plays a more important role in modulating basal MxA expression, thus contributing more significantly to innate immune response against viral infections that suppress endogenous IFN-alpha and IFN-beta induction such as SARS coronavirus.
7.
Increased sensitivity of SARS-coronavirus to a combination of human type I and type II interferons.
Scagnolari C, Vicenzi E, Bellomi F, Stillitano MG, Pinna D, Poli G, Clementi M, Dianzani F, Antonelli G. Antivir Ther. 2004 Dec;9(6):1003-11. PMID: 15651759
There is currently an urgent need to identify effective antiviral agents that will prevent and treat severe acute respiratory syndrome coronavirus (SARS-CoV) infection. In this study, we have investigated and compared the antiviral effect of different interferons (IFNs) on SARS-CoV replication in the epithelial kidney monkey Vero cell line. The results showed that SARS-CoV grown in Vero cells is moderately sensitive to IFN-beta and only weakly sensitive to IFN-alpha and IFN-gamma, in comparison to other IFN-sensitive viruses, such as those for encephalomyocarditis, vesicular stomatitis and Newcastle disease. Simultaneous incubation of Vero cells with IFN-beta and IFN-gamma indicated that they may act synergistically against SARS-CoV replication. The IFN-induced MxA protein was detected in the IFN-treated Vero cells. The data, however, suggest that the antiviral activity of IFN against SARS-CoV virus is independent of MxA expression. 
 
 8.
Multi-tiered screening and diagnosis strategy for COVID-19: a model for sustainable testing capacity in response to pandemic.
Pulia MS, O'Brien TP, Hou PC, Schuman A, Sambursky R. Ann Med. 2020 May 14:1-8. doi: 10.1080/07853890.2020.1763449. Online ahead of print. PMID: 32370561
Coronavirus disease 2019 (COVID-19), caused by novel enveloped single stranded RNA coronavirus (SARS-CoV-2), is responsible for an ongoing global pandemic. While other countries deployed widespread testing as an early mitigation strategy, the U.S. experienced delays in development and deployment of organism identification assays. As such, there is uncertainty surrounding disease burden and community spread, severely hampering containment efforts. COVID-19 illuminates the need for a tiered diagnostic approach to rapidly identify clinically significant infections and reduce disease spread. Without the ability to efficiently screen patients, hospitals are overwhelmed, potentially delaying treatment for other emergencies. A multi-tiered, diagnostic strategy incorporating a rapid host immune response assay as a screening test, molecular confirmatory testing and rapid IgM/IgG testing to assess benefit from quarantine/further testing and provide information on population exposure/herd immunity would efficiently evaluate potential COVID-19 patients. Triaging patients within minutes with a fingerstick rather than hours/days after an invasive swab is critical to pandemic response as reliance on the existing strategy is limited by assay accuracy, time to results, and testing capacity. Early screening and triage is achievable from the outset of a pandemic with point-of-care host immune response testing which will improve response time to clinical and public health actions.Key messages Delayed testing deployment has led to uncertainty surrounding overall disease burden and community spread, severely hampering public health containment and healthcare system preparation efforts.A multi-tiered testing strategy incorporating rapid, host immune point-of-care tests can be used now and for future pandemic planning by effectively identifying patients at risk of disease thereby facilitating quarantine earlier in the progression of the outbreak during the weeks and months it can take for pathogen specific confirmatory tests to be developed, validated and manufactured in sufficient quantities.The ability to triage patients at the point of care and support the guidance of medical and therapeutic decisions, for viral isolation or confirmatory testing or for appropriate treatment of COVID-19 and/or bacterial infections, is a critical component to our national pandemic response and there is an urgent need to implement the proposed strategy to combat the current outbreak. 
 
 9.
Association of SARS susceptibility with single nucleic acid polymorphisms of OAS1 and MxA genes: a case-control study.
He J, Feng D, de Vlas SJ, Wang H, Fontanet A, Zhang P, Plancoulaine S, Tang F, Zhan L, Yang H, Wang T, Richardus JH, Habbema JD, Cao W. BMC Infect Dis. 2006 Jul 6;6:106. doi: 10.1186/1471-2334-6-106. PMID: 16824203 Free PMC article. Background: Host genetic factors may play a role in susceptibility and resistance to SARS associated coronavirus (SARS-CoV) infection. The study was carried out to investigate the association between the genetic polymorphisms of 2',5'-oligoadenylate synthetase 1 (OAS1) gene as well as myxovirus resistance 1 (MxA) gene and susceptibility to SARS in Chinese Han population.
Methods: A hospital-based case-control study was conducted. A collective of 66 SARS cases and 64 close contact uninfected controls were enrolled in this study. End point real time polymerase chain reaction (PCR) and PCR-based Restriction Fragment Length Polymorphism (RFLP) analysis were used to detect the single nucleic polymorphisms (SNPs) in OAS1 and MxA genes. Information on other factors associated with SARS infection was collected using a pre-tested questionnaire. Univariate and multivariate logistic analyses were conducted.
Results: One polymorphism in the 3'-untranslated region (3'-UTR) of the OAS1 gene was associated with SARS infection. Compared to AA genotype, AG and GG genotypes were found associated with a protective effect on SARS infection with ORs (95% CI) of 0.42 (0.20-0.89) and 0.30 (0.09-0.97), respectively. Also, a GT genotype at position 88 in the MxA gene promoter was associated with increased susceptibility to SARS infection compared to a GG genotype (OR = 3.06, 95% CI: 1.25-7.50). The associations of AG genotype in OAS1 and GT genotype in MxA remained significant in multivariate analyses after adjusting for SARS protective measures (OR = 0.38, 95% CI: 0.14-0.98 and OR = 3.22, 95% CI: 1.13-9.18, respectively).
Conclusion: SNPs in the OAS1 3'-UTR and MxA promoter region appear associated with host susceptibility to SARS in Chinese Han population.

10. 
Severe acute respiratory syndrome coronavirus fails to activate cytokine-mediated innate immune responses in cultured human monocyte-derived dendritic cells.
Ziegler T, Matikainen S, Rönkkö E, Osterlund P, Sillanpää M, Sirén J, Fagerlund R, Immonen M, Melén K, Julkunen I. J Virol. 2005 Nov;79(21):13800-5. doi: 10.1128/JVI.79.21.13800-13805.2005. PMID: 16227300 Free PMC article.
Activation of host innate immune responses was studied in severe acute respiratory syndrome coronavirus (SCV)-infected human A549 lung epithelial cells, macrophages, and dendritic cells (DCs). In all cell types, SCV-specific subgenomic mRNAs were seen, whereas no expression of SCV proteins was found. No induction of cytokine genes (alpha interferon [IFN-alpha], IFN-beta, interleukin-28A/B [IL-28A/B], IL-29, tumor necrosis factor alpha, CCL5, or CXCL10) or IFN-alpha/beta-induced MxA gene was seen in SCV-infected A549 cells, macrophages, or DCs. SCV also failed to induce DC maturation (CD86 expression) or enhance major histocompatibility complex class II expression. Our data strongly suggest that SCV fails to activate host cell cytokine gene expression in human macrophages and DCs.
 

Inhibition of cytokine gene expression and induction of chemokine genes in non-lymphatic cells infected with SARS coronavirus.
Spiegel M, Weber F. Virol J. 2006 Mar 29;3:17. doi: 10.1186/1743-422X-3-17. PMID: 16571117 Free PMC article.
Background: SARS coronavirus (SARS-CoV) is the etiologic agent of the severe acute respiratory syndrome. SARS-CoV mainly infects tissues of non-lymphatic origin, and the cytokine profile of those cells can determine the course of disease. Here, we investigated the cytokine response of two human non-lymphatic cell lines, Caco-2 and HEK 293, which are fully permissive for SARS-CoV.
Results: A comparison with established cytokine-inducing viruses revealed that SARS-CoV only weakly triggered a cytokine response. In particular, SARS-CoV did not activate significant transcription of the interferons IFN-alpha, IFN-beta, IFN-lambda1, IFN-lambda2/3, as well as of the interferon-induced antiviral genes ISG56 and MxA, the chemokine RANTES and the interleukine IL-6. Interestingly, however, SARS-CoV strongly induced the chemokines IP-10 and IL-8 in the colon carcinoma cell line Caco-2, but not in the embryonic kidney cell line 293.
Conclusion: Our data indicate that SARS-CoV suppresses the antiviral cytokine system of non-immune cells to a large extent, thus buying time for dissemination in the host. However, synthesis of IP-10 and IL-8, which are established markers for acute-stage SARS, escapes the virus-induced silencing at least in some cell types. Therefore, the progressive infiltration of immune cells into the infected lungs observed in SARS patients could be due to the production of these chemokines by the infected tissue cells.
 

OAS-RNAse L, Coronavirus, haku (10)

https://pubmed.ncbi.nlm.nih.gov/?term=OAS-RNase+L%2C++coronavirus

OAS-RNaasi L välittää DNA-metylaatiolääkkeiden sytotoksisuutta.

OAS-RNase L Innate Immune Pathway Mediates the Cytotoxicity of a DNA-demethylating Drug
Affiliations
DOI: 10.1073/pnas.1815071116
Free PMC article
Abstract
Drugs that reverse epigenetic silencing, such as the DNA methyltransferase inhibitor (DNMTi) 5-azacytidine (AZA), have profound effects on transcription and tumor cell survival. AZA is an approved drug for myelodysplastic syndromes and acute myeloid leukemia, and is under investigation for different solid malignant tumors. AZA treatment generates self, double-stranded RNA (dsRNA), transcribed from hypomethylated repetitive elements. Self dsRNA accumulation in DNMTi-treated cells leads to type I IFN production and IFN-stimulated gene expression. Here we report that cell death in response to AZA treatment occurs through the 2',5'-oligoadenylate synthetase (OAS)-RNase L pathway.

  OASs are IFN-induced enzymes that synthesize the RNase L activator 2-5A in response to dsRNA. Cells deficient in RNase L or OAS1 to 3 are highly resistant to AZA, as are wild-type cells treated with a small-molecule inhibitor of RNase L. A small-molecule inhibitor of c-Jun NH2-terminal kinases (JNKs) also antagonizes RNase L-dependent cell death in response to AZA, consistent with a role for JNK in RNase L-induced apoptosis.

 In contrast, the rates of AZA-induced and RNase L-dependent cell death were increased by transfection of 2-5A, by deficiencies in ADAR1 (which edits and destabilizes dsRNA), PDE12 or AKAP7 (which degrade 2-5A), or by ionizing radiation (which induces IFN-dependent signaling).
Finally, OAS1 expression correlates with AZA sensitivity in the NCI-60 set of tumor cell lines, suggesting that the level of OAS1 can be a biomarker for predicting AZA sensitivity of tumor cells. These studies may eventually lead to pharmacologic strategies for regulating the antitumor activity and toxicity of AZA and related drugs.
Keywords: 5-azacytidine; DNA methyltransferase inhibitor; OAS; RNase L; innate immunity.


2016 Feb 23;113(8):2241-6.
doi: 10.1073/pnas.1519657113. Epub 2016 Feb 8.
Activation of RNase L Is Dependent on OAS3 Expression During Infection With Diverse Human Viruses
DOI: 10.1073/pnas.1519657113 
The 2′,5′-oligoadenylate (2-5A) synthetase (OAS)–RNase L system is an IFN-induced antiviral pathway. RNase L activity depends on 2-5A, synthesized by OAS. Although all three enzymatically active OAS proteins in humans—OAS1, OAS2, and OAS3—synthesize 2-5A upon binding dsRNA, it is unclear which are responsible for RNase L activation during viral infection. We used clustered regularly interspaced short palindromic repeats (CRISPR)–CRISPR-associated protein-9 nuclease (Cas9) technology to engineer human A549-derived cell lines in which each of the OAS genes or RNase L is knocked out. Upon transfection with poly(rI):poly(rC), a synthetic surrogate for viral dsRNA, or infection with each of four viruses from different groups (West Nile virus, Sindbis virus, influenza virus, or vaccinia virus), OAS1-KO and OAS2-KO cells synthesized amounts of 2-5A similar to those synthesized in parental wild-type cells, causing RNase L activation as assessed by rRNA degradation. In contrast, OAS3-KO cells synthesized minimal 2-5A, and rRNA remained intact, similar to infected RNase L-KO cells. All four viruses replicated to higher titers in OAS3-KO or RNase L-KO A549 cells than in parental, OAS1-KO, or OAS2-KO cells, demonstrating the antiviral effects of OAS3. OAS3 displayed a higher affinity for dsRNA in intact cells than either OAS1 or OAS2, consistent with its dominant role in RNase L activation. Finally, the requirement for OAS3 as the major OAS isoform responsible for RNase L activation was not restricted to A549 cells, because OAS3-KO cells derived from two other human cell lines also were deficient in RNase L activation. 
 
Figure 6.
An active RNase L dimer preferentially localizes to the polysome to form a complex with Dom34. (A) HeLa cells were transfected with a combination of pCMV-5×Myc-Dom34 (lanes 1–6) and either pCMV-5×Flag (lanes 1 and 4), pCMV-5×Flag-RNase L (lanes 2 and 5) or pCMV-5×Flag-RNase L Y312A (lanes 3 and 6). The cells were lysed in buffer B. The immunoprecipitates (lanes 4–6) and inputs (lanes 1–3, 10% of the amount immunoprecipitated) were analyzed by western blotting with the indicated antibodies. The leftmost five lanes, which analyzed 3-fold dilutions of total protein, show that the conditions used for western blotting are semi-quantitative. (B) The amount of immunoprecipitated 5×Myc-Dom34 protein in (A) was measured and normalized to the input 5×Myc-Dom34 protein, and either immunoprecipitated 5×Flag-RNase L wt or RNase L Y312A. Dom34 protein level in pCMV-5×Flag-RNase L wt-transfected cells (lane 5) was defined as 100% and Dom34 protein level in pCMV-5×Flag-RNase L Y312A-transfected cells (lane 6) was represented. (C) HeLa/5×Flag-RNase L cells were transfected with 2–5A using Neon™ Transfection System. At 6 h after 2–5A electroporation, the cells were harvested and lysed in buffer G. The cell lysate was fractionated by sucrose gradients. 5×Flag-RNase L in the polysomal fractions and the input (0.5% of the amount loaded onto sucrose gradients) was analyzed by western blotting with an anti-Flag antibody. (D) The amount of endogenous 5×Flag-tagged RNase L protein in (C) was measured and normalized to the input. RNase L protein levels in mock cells were set to 1 and fold-changes were indicated (mean ± SEM, n = 3). (E) Model for the Dom34-mediated decay of exogenous mRNA (see Discussion).
 

OAS/RNase L systgeemi. Primäärinen virus vaste dsRNA viruksen havaitsemisessa

 Sitaatti: 
"Also the 2´-5´ oligoadenylate synthetase (OAS)  family members are triggered by viral dsRNA (2011). In the dsRNA-bound state they synthetize short chains of 2´-5´ oligoadenylates that activate the latent RNase L. RNase L then cleaves virus and host ssRNAs, predominantly at single stranded UA and UU dinucleotides (Wreschner 1981). Interestingly, the small 3´-monophosphorylated cleavage products of RNase L are recognized by the PRRs RIG-1 and MDA5, thus amplifying the IFN response in an infection-dependent manner(Malathi , 2007). polymorphism of the OAS-1 gene might affect susceptibility  to SARS-CoV (Hamano, 2005), but to our knowledge, there is no direct data on antiviral effects of the OAS/RNase L system on human coronaviruses.  For the mouse coronavirus MHV-A59, however, it was shown that mutants deficient in the ns2 gene are highly sensitive against RNaSE l 8zHAO 2012). "
Kindler E e al.  INTERACTION OF SARS AND MERS CORONAVIRUSES WITH THE ANTIVIRAL INTERFERON RESPONSE. 
http://dx.doi.org/10.1016/bs.aivir.2016.08.006 http://dx.doi.org/10.1016/bs.aivir.2016.08.006

  • Etsin lisätietoa OAS/RNase L  tiestä yleensä.

https://pubmed.ncbi.nlm.nih.gov/?term=OAS%2FRNase+L+system

Drappier M, Michiels T. Curr Opin Virol. 2015 Dec;15:19-26. doi: 10.1016/j.coviro.2015.07.002. Epub 2015 Jul 29. PMID: 26231767 Free PMC article. Review.

The OAS/RNase L system was one of the first characterized interferon effector pathways. It relies on the synthesis, by oligoadenylate synthetases (OAS), of short oligonucleotides that act as second messengers to activate the latent cellular RNase L. Viruses have developed diverse strategies to escape its antiviral effects. This underscores the importance of the OAS/RNase L pathway in antiviral defenses. Viral proteins such as the NS1 protein of Influenza virus A act upstream of the pathway while other viral proteins such as Theiler's virus L* protein act downstream. The diversity of escape strategies used by viruses likely stems from their relative susceptibility to OAS/RNase L and other antiviral pathways, which may depend on their host and cellular tropism.
 
Li Y, Dong B, Wei Z, Silverman RH, Weiss SR. mBio. 2019 Nov 12;10(6):e02414-19. doi: 10.1128/mBio.02414-19. PMID: 31719180 Free PMC article.
Bats are reservoirs for many RNA viruses that are highly pathogenic in humans yet relatively apathogenic in the natural host. It has been suggested that differences in innate immunity are responsible. The antiviral OAS-RNase L pathway is well characterized in humans, but there is little known about its activation and antiviral activity in bats. During infection, OASs, upon sensing double-stranded RNA (dsRNA), produce 2'-5' oligoadenylates (2-5A), leading to activation of RNase L which degrades viral and host RNA, limiting viral replication. Humans encode three active OASs (OAS1 to -3). Analysis of the Egyptian Rousette bat genome combined with mRNA sequencing from bat RoNi/7 cells revealed three homologous OAS proteins. Interferon alpha treatment or viral infection induced all three OAS mRNAs, but RNase L mRNA is constitutively expressed.
 Sindbis virus (SINV) or vaccinia virus (VACVΔE3L) infection of wild-type (WT) or OAS1-KO (knockout), OAS2-KO, or MAVS-KO RoNi/7 cells, but not RNase L-KO or OAS3-KO cells, induces robust RNase L activation. SINV replication is 100- to 200-fold higher in the absence of RNase L or OAS3 than in WT cells. However, MAVS-KO had no detectable effect on RNA degradation or replication. Thus, in RoNi/7 bat cells, as in human cells, activation of RNase L during infection and its antiviral activity are dependent primarily on OAS3 while MAVS signaling is not required for the activation of RNase L and restriction of infection. 
Our findings indicate that OAS proteins serve as pattern recognition receptors (PRRs) to recognize viral dsRNA and that this pathway is a primary response to virus rather than a secondary effect of interferon signaling. 
IMPORTANCE Many RNA viruses that are highly pathogenic in humans are relatively apathogenic in their bat reservoirs, making it important to compare innate immune responses in bats to those well characterized in humans. One such antiviral response is the OAS-RNase L pathway. OASs, upon sensing dsRNA, produce 2-5A, leading to activation of RNase L which degrades viral and host RNA, limiting viral replication. 
Analysis of Egyptian Rousette bat sequences revealed three OAS genes expressing OAS1, OAS2, and OAS3 proteins. Interferon treatment or viral infection induces all three bat OAS mRNAs. In these bat cells as in human cells, RNase L activation and its antiviral activity are dependent primarily on OAS3 while MAVS signaling is not required.
 Importantly, our findings indicate the OAS-RNase L system is a primary response to virus rather than a secondary effect of interferon signaling and therefore can be activated early in infection or while interferon signaling is antagonized.
 
Rong E, Wang X, Chen H, Yang C, Hu J, Liu W, Wang Z, Chen X, Zheng H, Pu J, Sun H, Smith J, Burt DW, Liu J, Li N, Huang Y. Front Immunol. 2018 Jun 20;9:1398. doi: 10.3389/fimmu.2018.01398. eCollection 2018. PMID: 29973937 Free PMC article.
Abstract
Host cells develop
(*) the OAS/RNase L [2'-5'-oligoadenylate synthetase (OAS)/ribonuclease L] system to degrade cellular and viral RNA, and/or
(**) the OASL/RIG-I (2'-5'-OAS like/retinoic acid inducible protein I) system to enhance RIG-I-mediated IFN induction,
 thus providing the first line of defense against viral infection.
( *)The 2'-5'-OAS-like (OASL) protein may activate the OAS/RNase L system using its typical OAS-like domain (OLD) 
or(**)mimic the K63-linked pUb to enhance antiviral activity of the OASL/RIG-I system using its two tandem ubiquitin-like domains (UBLs).
We first describe that divergent avian (duck and ostrich) OASL inhibit the replication of a broad range of RNA viruses by activating and magnifying the OAS/RNase L pathway(***) in a UBL-dependent manner.
This is in sharp contrast to mammalian enzymatic OASL, which activates and magnifies (*) the OAS/RNase L pathway   in a UBL-independent manner, similar to 2'-5'-oligoadenylate synthetase 1 (OAS1).
 We further show that both avian and mammalian OASL can reversibly exchange to activate and magnify the OAS/RNase L and OASL/RIG-I system by introducing only three key residues, suggesting that ancient OASL possess 2-5A [px5'A(2'p5'A)n; x = 1-3; n ≥ 2] activity and has functionally switched to the OASL/RIG-I pathway recently. Our findings indicate the molecular mechanisms involved in the switching of avian and mammalian OASL molecules to activate and enhance the OAS/RNase L and OASL/RIG-I pathways in response to infection by RNA viruses.
 
Schwartz SL, Conn GL. Wiley Interdiscip Rev RNA. 2019 Jul;10(4):e1534. doi: 10.1002/wrna.1534. Epub 2019 Apr 15. PMID: 30989826 Review.
The innate immune system is a broad collection of critical intra- and extra-cellular processes that limit the infectivity of diverse pathogens. The 2'-5'-oligoadenylate synthetase (OAS) family of enzymes are important sensors of cytosolic double-stranded RNA (dsRNA) that play a critical role in limiting viral infection by activating the latent ribonuclease (RNase L) to halt viral replication and establish an antiviral state. 
Attesting to the importance of the OAS/RNase L pathway, diverse viruses have developed numerous distinct strategies to evade the effects of OAS activation. How OAS proteins are regulated by viral or cellular RNAs is not fully understood but several recent studies have provided important new insights into the molecular mechanisms of OAS activation by dsRNA. Other studies have revealed unanticipated features of RNA sequence and structure that strongly enhance activation of at least one OAS family member. While these discoveries represent important advances, they also underscore the fact that much remains to be learned about RNA-mediated regulation of the OAS/RNase L pathway.  
 In particular, defining the full complement of RNA molecular signatures that activate OAS is essential to our understanding of how these proteins maximize their protective role against pathogens while still accurately discriminating host molecules to avoid inadvertent activation by cellular RNAs. A more complete knowledge of OAS regulation may also serve as a foundation for the development of novel antiviral therapeutic strategies and lead the way to a deeper understanding of currently unappreciated cellular functions of the OAS/RNase L pathway in the absence of infection. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications Translation > Translation Regulation. 
 
Lamers MM, van den Hoogen BG, Haagmans BL. Front Immunol. 2019 Jul 25;10:1763. doi: 10.3389/fimmu.2019.01763. eCollection 2019. PMID: 31404141 Free PMC article. Review.
Specialized receptors that recognize molecular patterns such as double stranded RNA duplexes-indicative of viral replication-are potent triggers of the innate immune system.
 Although their activation is beneficial during viral infection, RNA transcribed from endogenous mobile genetic elements may also act as ligands potentially causing autoimmunity. Recent advances indicate that the adenosine deaminase ADAR1 through RNA editing is involved in dampening the canonical antiviral RIG-I-like receptor-, PKR-, and OAS-RNAse L pathways to prevent autoimmunity. However, this inhibitory effect must be overcome during viral infections. In this review we discuss ADAR1's critical role in balancing immune activation and self-tolerance.
Chakrabarti A, Banerjee S, Franchi L, Loo YM, Gale M Jr, Núñez G, Silverman RH. Cell Host Microbe. 2015 Apr 8;17(4):466-77. doi: 10.1016/j.chom.2015.02.010. Epub 2015 Mar 26. PMID: 25816776 Free PMC article.
The NLRP3 inflammasome assembles in response to danger signals, triggering self-cleavage of procaspase-1 and production of the proinflammatory cytokine IL-1β. 
 Although virus infection activates the NLRP3 inflammasome, the underlying events remain incompletely understood. We report that virus activation of the NLRP3 inflammasome involves the 2',5'-oligoadenylate (2-5A) synthetase(OAS)/RNase L system, a component of the interferon-induced antiviral response that senses double-stranded RNA and activates endoribonuclease RNase L to cleave viral and cellular RNAs. The absence of RNase L reduces IL-1β production in influenza A virus-infected mice. RNA cleavage products generated by RNase L enhance IL-1β production but require the presence of 2',3'-cyclic phosphorylated termini characteristic of RNase L activity. Additionally, these cleavage products stimulate NLRP3 complex formation with the DExD/H-box helicase, DHX33, and mitochondrial adaptor protein, MAVS, which are each required for effective NLRP3 inflammasome activation. Thus, RNA cleavage events catalyzed by RNase L are required for optimal inflammasome activation during viral infections.