Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Viruses ; 15(7)2023 07 13.
Article in English | MEDLINE | ID: mdl-37515225

ABSTRACT

Viral replication often depends on RNA maturation and degradation processes catalyzed by viral ribonucleases, which are therefore candidate targets for antiviral drugs. Here, we synthesized and studied the antiviral properties of a novel nitrocatechol compound (1c) and other analogs that are structurally related to the catechol derivative dynasore. Interestingly, compound 1c strongly inhibited two DEDD box viral ribonucleases, HIV-1 RNase H and SARS-CoV-2 nsp14 3'-to-5' exoribonuclease (ExoN). While 1c inhibited SARS-CoV-2 ExoN activity, it did not interfere with the mRNA methyltransferase activity of nsp14. In silico molecular docking placed compound 1c in the catalytic pocket of the ExoN domain of nsp14. Finally, 1c inhibited SARS-CoV-2 replication but had no toxicity to human lung adenocarcinoma cells. Given its simple chemical synthesis from easily available starting materials, these results suggest that 1c might be a lead compound for the design of new antiviral compounds that target coronavirus nsp14 ExoN and other viral ribonucleases.


Subject(s)
COVID-19 , HIV-1 , Humans , SARS-CoV-2/genetics , Exoribonucleases/genetics , HIV-1/genetics , Molecular Docking Simulation , Antiviral Agents/pharmacology , Virus Replication , Catechols/pharmacology , Ribonuclease H/pharmacology , Viral Nonstructural Proteins/genetics , RNA, Viral/genetics
2.
Cell Rep ; 36(12): 109747, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34551289

ABSTRACT

PBRM1, a subunit of the PBAF coactivator complex that transcription factors use to activate target genes, is genetically inactivated in almost all clear cell renal cell cancers (RCCs). Using unbiased proteomic analyses, we find that PAX8, a master transcription factor driver of proximal tubule epithelial fates, recruits PBRM1/PBAF. Reverse analyses of the PAX8 interactome confirm recruitment specifically of PBRM1/PBAF and not functionally similar BAF. More conspicuous in the PAX8 hub in RCC cells, however, are corepressors, which functionally oppose coactivators. Accordingly, key PAX8 target genes are repressed in RCC versus normal kidneys, with the loss of histone lysine-27 acetylation, but intact lysine-4 trimethylation, activation marks. Re-introduction of PBRM1, or depletion of opposing corepressors using siRNA or drugs, redress coregulator imbalance and release RCC cells to terminal epithelial fates. These mechanisms thus explain RCC resemblance to the proximal tubule lineage but with suppression of the late-epithelial program that normally terminates lineage-precursor proliferation.


Subject(s)
Carcinoma, Renal Cell/pathology , Cell Differentiation , DNA-Binding Proteins/metabolism , Kidney Tubules, Proximal/metabolism , PAX8 Transcription Factor/metabolism , Transcription Factors/metabolism , Animals , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Histones/metabolism , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Tubules, Proximal/cytology , Male , Mice , Mice, Nude , Mutagenesis , PAX8 Transcription Factor/genetics , Protein Interaction Maps , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcriptional Activation , Transplantation, Heterologous
3.
Leukemia ; 32(8): 1751-1761, 2018 08.
Article in English | MEDLINE | ID: mdl-29795413

ABSTRACT

Somatic mutations in TET2 are common in myelodysplastic syndromes (MDS), myeloproliferative, and overlap syndromes. TET2 mutant (TET2MT) clones are also found in asymptomatic elderly individuals, a condition referred to as clonal hematopoiesis of indeterminate potential (CHIP). In various entities of TET2MT neoplasia, we examined the phenotype in relation to the strata of TET2 hits within the clonal hierarchy. Using deep sequencing, 1781 mutations were found in 1205 of 4930 patients; 40% of mutant cases were biallelic. Hierarchical analysis revealed that of TET2MT cases >40% were ancestral, e.g., representing 8% of MDS. Higher (earlier) TET2 lesion rank within the clonal hierarchy (greater clonal burden) was associated with impaired survival. Moreover, MDS driven by ancestral TET2MT is likely derived from TET2MT CHIP with a penetrance of ~1%. Following ancestral TET2 mutations, individual disease course is determined by secondary hits. Using multidimensional analyses, we demonstrate how hits following the TET2 founder defect induces phenotypic shifts toward dysplasia, myeloproliferation, or progression to AML. In summary, TET2MT CHIP-derived MDS is a subclass of MDS that is distinct from de novo disease.


Subject(s)
Clone Cells/pathology , DNA-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Proto-Oncogene Proteins/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Differentiation , Clonal Evolution , Clone Cells/metabolism , Dioxygenases , Disease Progression , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Prognosis
4.
PLoS Pathog ; 14(4): e1006989, 2018 04.
Article in English | MEDLINE | ID: mdl-29652922

ABSTRACT

The OAS/RNase L pathway is one of the best-characterized effector pathways of the IFN antiviral response. It inhibits the replication of many viruses and ultimately promotes apoptosis of infected cells, contributing to the control of virus spread. However, viruses have evolved a range of escape strategies that act against different steps in the pathway. Here we unraveled a novel escape strategy involving Theiler's murine encephalomyelitis virus (TMEV) L* protein. Previously we found that L* was the first viral protein binding directly RNase L. Our current data show that L* binds the ankyrin repeats R1 and R2 of RNase L and inhibits 2'-5' oligoadenylates (2-5A) binding to RNase L. Thereby, L* prevents dimerization and oligomerization of RNase L in response to 2-5A. Using chimeric mouse hepatitis virus (MHV) expressing TMEV L*, we showed that L* efficiently inhibits RNase L in vivo. Interestingly, those data show that L* can functionally substitute for the MHV-encoded phosphodiesterase ns2, which acts upstream of L* in the OAS/RNase L pathway, by degrading 2-5A.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , Adenine Nucleotides/metabolism , Endoribonucleases/antagonists & inhibitors , Murine hepatitis virus/physiology , Oligoribonucleotides/metabolism , Theilovirus/metabolism , Viral Proteins/metabolism , Animals , Antiviral Agents/metabolism , Endoribonucleases/physiology , HeLa Cells , Hepatitis, Viral, Animal/metabolism , Hepatitis, Viral, Animal/virology , Host-Pathogen Interactions , Humans , Mice
5.
mBio ; 5(2): e00856-14, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24570368

ABSTRACT

The interferon (IFN)-inducible antiviral state is mediated in part by the 2',5'-oligoadenylate (2-5A) synthetase (OAS)/RNase L system. 2-5A, produced from ATP by OAS proteins in response to viral double-stranded RNA, binds to and activates RNase L. RNase L restricts viral infections by degrading viral and cellular RNA, inducing autophagy and apoptosis, and producing RNA degradation products that amplify production of type I interferons (IFNs) through RIG-I-like receptors. However, the effects of the OAS/RNase L pathway on IFN induction in different cell types that vary in basal levels of these proteins have not been previously reported. Here we report higher basal expression of both RNase L and OAS in mouse macrophages in comparison to mouse embryonic fibroblasts (MEFs). In MEFs, RNase L gene knockout decreased induction of IFN-ß by encephalomyocarditis virus infection or poly(rI):poly(rC) (pIC) transfection. In contrast, in macrophages, RNase L deletion increased (rather than decreased) induction of IFN-ß by virus or pIC. RNA damage from RNase L in virus-infected macrophages is likely responsible for reducing IFN-ß production. Similarly, direct activation of RNase L by transfection with 2-5A induced IFN-ß in MEFs but not in macrophages. Also, viral infection or pIC transfection caused RNase L-dependent apoptosis of macrophages but not of MEFs. Our results suggest that cell-type-specific differences in basal levels of OAS and RNase L are determinants of IFN-ß induction that could affect tissue protection and survival during viral infections. IMPORTANCE Type I interferons (IFNs) such as IFN-ß are essential antiviral cytokines that are often required for animal survival following infections by highly pathogenic viruses. Therefore, host factors that regulate type I IFN production are critically important for animal and human health. Previously we reported that the OAS/RNase L pathway amplifies antiviral innate immunity by enhancing IFN-ß production in mouse embryonic fibroblasts and in virus-infected mice. Here we report that high basal levels of OAS/RNase L in macrophages reduce, rather than increase, virus induction of IFN-ß. RNA damage and apoptosis caused by RNase L were the likely reasons for the decreased IFN-ß production in virus-infected macrophages. Our studies suggest that during viral infections, the OAS/RNase L pathway can either enhance or suppress IFN production, depending on the cell type. IFN regulation by RNase L is suggested to contribute to tissue protection and survival during viral infections.


Subject(s)
2',5'-Oligoadenylate Synthetase/immunology , Encephalomyocarditis virus/immunology , Endoribonucleases/immunology , Fibroblasts/immunology , Interferon-beta/immunology , Interferon-beta/metabolism , Macrophages/immunology , 2',5'-Oligoadenylate Synthetase/metabolism , Animals , Endoribonucleases/metabolism , Fibroblasts/virology , Macrophages/virology , Mice
6.
Mol Cell ; 53(2): 221-34, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24462203

ABSTRACT

RNase L is an ankyrin repeat domain-containing dual endoribonuclease-pseudokinase that is activated by unusual 2,'5'-oligoadenylate (2-5A) second messengers and which impedes viral infections in higher vertebrates. Despite its importance in interferon-regulated antiviral innate immunity, relatively little is known about its precise mechanism of action. Here we present a functional characterization of 2.5 Å and 3.25 Å X-ray crystal and small-angle X-ray scattering structures of RNase L bound to a natural 2-5A activator with and without ADP or the nonhydrolysable ATP mimetic AMP-PNP. These studies reveal how recognition of 2-5A through interactions with the ankyrin repeat domain and the pseudokinase domain, together with nucleotide binding, imposes a rigid intertwined dimer configuration that is essential for RNase catalytic and antiviral functions. The involvement of the pseudokinase domain of RNase L in 2-5A sensing, nucleotide binding, dimerization, and ribonuclease functions highlights the evolutionary adaptability of the eukaryotic protein kinase fold.


Subject(s)
Adenine Nucleotides/chemistry , Endoribonucleases/chemistry , Oligoribonucleotides/chemistry , Adenosine Diphosphate/chemistry , Adenylyl Imidodiphosphate/chemistry , Animals , Ankyrin Repeat , Binding Sites , Crystallography, X-Ray , Dimerization , Encephalomyocarditis virus , Endoribonucleases/genetics , Endoribonucleases/physiology , HeLa Cells , Humans , Models, Molecular , Mutagenesis, Site-Directed , Picornaviridae , Protein Structure, Tertiary , Scattering, Radiation , Structure-Activity Relationship , Sus scrofa
7.
J Immunol ; 191(5): 2637-46, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23913960

ABSTRACT

Interactions between cigarette smoke (CS) exposure and viral infection play an important role(s) in the pathogenesis of chronic obstructive pulmonary disease and a variety of other disorders. A variety of lines of evidence suggest that this interaction induces exaggerated inflammatory, cytokine, and tissue remodeling responses. We hypothesized that the 2'-5' oligoadenylate synthetase (OAS)/RNase L system, an innate immune antiviral pathway, plays an important role in the pathogenesis of these exaggerated responses. To test this hypothesis, we characterize the activation of 2'-5' OAS in lungs from mice exposed to CS and viral pathogen-associated molecular patterns (PAMPs)/live virus, alone and in combination. We also evaluated the inflammatory and remodeling responses induced by CS and virus/viral PAMPs in lungs from RNase L null and wild-type mice. These studies demonstrate that CS and viral PAMPs/live virus interact in a synergistic manner to stimulate the production of select OAS moieties. They also demonstrate that RNase L plays a critical role in the pathogenesis of the exaggerated inflammatory, fibrotic, emphysematous, apoptotic, TGF-ß1, and type I IFN responses induced by CS plus virus/viral PAMP in combination. These studies demonstrate that CS is an important regulator of antiviral innate immunity, highlight novel roles of RNase L in CS plus virus induced inflammation, tissue remodeling, apoptosis, and cytokine elaboration and highlight pathways that may be operative in chronic obstructive pulmonary disease and mechanistically related disorders.


Subject(s)
Airway Remodeling/physiology , Endoribonucleases/metabolism , Inflammation/enzymology , Orthomyxoviridae Infections/complications , Tobacco Smoke Pollution/adverse effects , Animals , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Inflammation/etiology , Inflammation/pathology , Influenza A virus , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/enzymology , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/pathology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
8.
PLoS Pathog ; 9(6): e1003474, 2013.
Article in English | MEDLINE | ID: mdl-23825954

ABSTRACT

Theiler's virus is a neurotropic picornavirus responsible for chronic infections of the central nervous system. The establishment of a persistent infection and the subsequent demyelinating disease triggered by the virus depend on the expression of L*, a viral accessory protein encoded by an alternative open reading frame of the virus. We discovered that L* potently inhibits the interferon-inducible OAS/RNase L pathway. The antagonism of RNase L by L* was particularly prominent in macrophages where baseline oligoadenylate synthetase (OAS) and RNase L expression levels are elevated, but was detectable in fibroblasts after IFN pretreatment. L* mutations significantly affected Theiler's virus replication in primary macrophages derived from wild-type but not from RNase L-deficient mice. L* counteracted the OAS/RNase L pathway through direct interaction with the ankyrin domain of RNase L, resulting in the inhibition of this enzyme. Interestingly, RNase L inhibition was species-specific as Theiler's virus L* protein blocked murine RNase L but not human RNase L or RNase L of other mammals or birds. Direct RNase L inhibition by L* and species specificity were confirmed in an in vitro assay performed with purified proteins. These results demonstrate a novel viral mechanism to elude the antiviral OAS/RNase L pathway. By targeting the effector enzyme of this antiviral pathway, L* potently inhibits RNase L, underscoring the importance of this enzyme in innate immunity against Theiler's virus.


Subject(s)
Cardiovirus Infections/metabolism , Endoribonucleases/antagonists & inhibitors , Immune Evasion/physiology , Immunity, Innate , Theilovirus/metabolism , Viral Proteins/metabolism , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/immunology , Cardiovirus Infections/pathology , Cell Line , Cricetinae , Endoribonucleases/genetics , Endoribonucleases/immunology , Endoribonucleases/metabolism , Humans , Mice , Mice, Mutant Strains , Protein Structure, Tertiary , Species Specificity , Theilovirus/genetics , Theilovirus/immunology , Viral Proteins/genetics , Viral Proteins/immunology
9.
J Virol ; 86(10): 5708-18, 2012 May.
Article in English | MEDLINE | ID: mdl-22379089

ABSTRACT

We previously showed that a noncoding subgenomic flavivirus RNA (sfRNA) is required for viral pathogenicity, as a mutant West Nile virus (WNV) deficient in sfRNA production replicated poorly in wild-type mice. To investigate the possible immunomodulatory or immune evasive functions of sfRNA, we utilized mice and cells deficient in elements of the type I interferon (IFN) response. Replication of the sfRNA mutant WNV was rescued in mice and cells lacking interferon regulatory factor 3 (IRF-3) and IRF-7 and in mice lacking the type I alpha/beta interferon receptor (IFNAR), suggesting a contribution for sfRNA in overcoming the antiviral response mediated by type I IFN. This was confirmed by demonstrating rescue of mutant virus replication in the presence of IFNAR neutralizing antibodies, greater sensitivity of mutant virus replication to IFN-α pretreatment, partial rescue of its infectivity in cells deficient in RNase L, and direct effects of transfected sfRNA on rescuing replication of unrelated Semliki Forest virus in cells pretreated with IFN-α. The results define a novel function of sfRNA in flavivirus pathogenesis via its contribution to viral evasion of the type I interferon response.


Subject(s)
Immune Evasion , Interferon Type I/immunology , RNA, Untranslated/immunology , RNA, Viral/immunology , West Nile Fever/immunology , West Nile virus/immunology , Animals , Cell Line , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Untranslated/genetics , RNA, Viral/genetics , Virulence , West Nile Fever/virology , West Nile virus/genetics , West Nile virus/pathogenicity
10.
Proc Natl Acad Sci U S A ; 109(15): E869-78, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-22315414

ABSTRACT

IRE1 couples endoplasmic reticulum unfolded protein load to RNA cleavage events that culminate in the sequence-specific splicing of the Xbp1 mRNA and in the regulated degradation of diverse membrane-bound mRNAs. We report on the identification of a small molecule inhibitor that attains its selectivity by forming an unusually stable Schiff base with lysine 907 in the IRE1 endonuclease domain, explained by solvent inaccessibility of the imine bond in the enzyme-inhibitor complex. The inhibitor (abbreviated 4µ8C) blocks substrate access to the active site of IRE1 and selectively inactivates both Xbp1 splicing and IRE1-mediated mRNA degradation. Surprisingly, inhibition of IRE1 endonuclease activity does not sensitize cells to the consequences of acute endoplasmic reticulum stress, but rather interferes with the expansion of secretory capacity. Thus, the chemical reactivity and sterics of a unique residue in the endonuclease active site of IRE1 can be exploited by selective inhibitors to interfere with protein secretion in pathological settings.


Subject(s)
Coumarins/pharmacology , Endoribonucleases/metabolism , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Splicing , Animals , Binding Sites , Coumarins/chemistry , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress , Endoribonucleases/antagonists & inhibitors , Humans , Lysine/metabolism , Membrane Proteins/antagonists & inhibitors , Mice , Protein Binding/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proteolysis/drug effects , RNA Splicing/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Factor X Transcription Factors , Ribonucleases/antagonists & inhibitors , Ribonucleases/metabolism , Schiff Bases/chemistry , Schiff Bases/metabolism , Secretory Pathway/drug effects , Transcription Factors/metabolism , X-Box Binding Protein 1
11.
RNA ; 18(1): 88-99, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22114318

ABSTRACT

Activation of RNase L endonuclease activity is part of the mammalian innate immune response to viral infection. The poliovirus RNA genome contains a sequence in its protein-coding region that can act as a competitive inhibitor of RNase L. Mutation, sequence, and functional analysis of this competitive inhibitor RNA (ciRNA) revealed that its activity depends on specific sequences, showed that a loop-loop hairpin interaction forms in the ciRNA, and suggested the presence of a loop E motif. These features lead to the hypothesis that the ciRNA's function is conferred in part by a specific three-dimensional folded RNA architecture. By using a combination of biophysical, mutational, and functional studies, we have mapped features of the three-dimensional architecture of the ciRNA in its unbound form. We show that the loop-loop interaction forms in the free ciRNA and affects the overall structure, perhaps forming long-range tertiary interactions with the loop E motif. Local tight RNA-RNA backbone packing occurs in parts of the structure, but the fold appears to be less stable than many other tightly packed RNAs. This feature may allow the ciRNA to accommodate the translocation of ribosomes and polymerase across this multifunctional region of the viral RNA but also to function as an RNase L inhibitor.


Subject(s)
Endoribonucleases/antagonists & inhibitors , RNA/chemistry , Animals , Base Sequence , Binding, Competitive , Endoribonucleases/chemistry , Endoribonucleases/genetics , Hot Temperature , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Nucleic Acid Denaturation , RNA/genetics , Scattering, Small Angle , X-Ray Diffraction
12.
J Biol Chem ; 286(30): 26319-26, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21636578

ABSTRACT

RNase L and RNA-dependent protein kinase (PKR) are effectors of the interferon antiviral response that share homology in their pseudokinase and protein kinase domains, respectively. Sunitinib is an orally available, ATP-competitive inhibitor of VEGF and PDGF receptors used clinically to suppress angiogenesis and tumor growth. Sunitinib also impacts IRE1, an endoplasmic reticulum protein involved in the unfolded protein response that is closely related to RNase L. Here, we report that sunitinib is a potent inhibitor of both RNase L and PKR with IC(50) values of 1.4 and 0.3 µM, respectively. In addition, flavonol activators of IRE1 inhibited RNase L. Sunitinib treatment of wild type (WT) mouse embryonic fibroblasts resulted in about a 12-fold increase in encephalomyocarditis virus titers. However, sunitinib had no effect on encephalomyocarditis virus growth in cells lacking both PKR and RNase L. Furthermore, oral delivery of sunitinib in WT mice resulted in 10-fold higher viral titers in heart tissues while suppressing by about 2-fold the IFN-ß levels. In contrast, sunitinib had no effect on viral titers in mice deficient in both RNase L and PKR. Also, sunitinib reduced mean survival times from 12 to 6 days in virus-infected WT mice while having no effect on survival of mice lacking both RNase L and PKR. Results indicate that sunitinib treatments prevent antiviral innate immune responses mediated by RNase L and PKR.


Subject(s)
Antineoplastic Agents/pharmacology , Cardiovirus Infections/immunology , Endoribonucleases/antagonists & inhibitors , Immunity, Innate/drug effects , Indoles/pharmacology , Membrane Proteins/immunology , Protein Serine-Threonine Kinases/immunology , Pyrroles/pharmacology , eIF-2 Kinase/antagonists & inhibitors , Animals , Cardiovirus Infections/enzymology , Cardiovirus Infections/genetics , Encephalomyocarditis virus , Endoribonucleases/genetics , Endoribonucleases/immunology , Endoribonucleases/metabolism , Humans , Immunity, Innate/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Sunitinib , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology
13.
J Interferon Cytokine Res ; 31(1): 49-57, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21190483

ABSTRACT

The interferon (IFN)-inducible 2'-5'-oligoadenylate synthetase (OAS)/RNase L pathway blocks infections by some types of viruses through cleavage of viral and cellular single-stranded RNA. Viruses induce type I IFNs that initiate signaling to the OAS genes. OAS proteins are pathogen recognition receptors for the viral pathogen-associated molecular pattern, double-stranded RNA. Double-stranded RNA activates OAS to produce p(x)5'A(2'p5'A)(n); x = 1-3; n > 2 (2-5A) from ATP. Upon binding 2-5A, RNase L is converted from an inactive monomer to a potently active dimeric endoribonuclease for single-stranded RNA. RNase L contains, from N- to C-terminus, a series of 9 ankyrin repeats, a linker, several protein kinase-like motifs, and a ribonuclease domain homologous to Ire1 (involved in the unfolded protein response). In the past few years, it has become increasingly apparent that RNase L and OAS contribute to innate immunity in many ways. For example, small RNA cleavage products produced by RNase L during viral infections can signal to the retinoic acid-inducible-I like receptors to amplify and perpetuate signaling to the IFN-ß gene. In addition, RNase L is now implicated in protecting the central nervous system against viral-induced demyelination. A role in tumor suppression was inferred by mapping of the RNase L gene to the hereditary prostate cancer 1 (HPC1) gene, which in turn led to discovery of the xenotropic murine leukemia-related virus. A broader role in innate immunity is suggested by involvement of RNase L in cytokine induction and endosomal pathways that suppress bacterial infections. These newly described findings about RNase L could eventually provide the basis for developing broad-spectrum antimicrobial drugs.


Subject(s)
Endoribonucleases/metabolism , Immunity, Innate , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Animals , Antigens, Surface/metabolism , Demyelinating Diseases/prevention & control , ELAV Proteins , ELAV-Like Protein 1 , Endoribonucleases/genetics , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Interferons/genetics , Interferons/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Protein Interaction Domains and Motifs , RNA, Double-Stranded/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Sequence Homology, Amino Acid , Virus Diseases/immunology , Virus Diseases/metabolism
14.
J Biol Chem ; 284(6): 3897-905, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19004836

ABSTRACT

Bacterial hyaluronan lyase enzymes are the major virulence factors that enable greater microbial ingress by cleaving hyaluronan (HA) polymers present predominantly in extracellular space of vertebrates. Based on the premise that effective inhibitors may bind to and stabilize HA thereby protecting it from degradation, here we investigated inhibitory activity of human hyaluronan-binding protein 1 (HABP1) on bacterial hyaluronidase because it is highly specific to HA and localized on the cell surface. Biochemical characterization revealed that HABP1 is a competitive inhibitor of Streptococcus pneumoniae hyaluronate lyase (SpnHL) with an IC50 value of 22 microm. This is thus the first report of an endogenous protein inhibitor that may be used during natural antibacterial defense. Our findings also support a novel multipronged mechanism for the high efficacy of HABP1-mediated inhibition based on structural modeling of enzyme, substrate, and inhibitor. Evidence from docking simulations and contact interface interactions showed that the inherent charge asymmetry of HABP1 plays a key role in the inhibitory activity. This novel role of HABP1 may pave the way for peptide inhibitors as alternatives to synthetic chemicals in antibacterial research.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Carrier Proteins/chemistry , Enzyme Inhibitors/chemistry , Hyaluronoglucosaminidase/antagonists & inhibitors , Mitochondrial Proteins/chemistry , Models, Molecular , Streptococcus pneumoniae/enzymology , Anti-Infective Agents/chemistry , Anti-Infective Agents/metabolism , Anti-Infective Agents/therapeutic use , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Enzyme Inhibitors/metabolism , Humans , Hyaluronoglucosaminidase/chemistry , Hyaluronoglucosaminidase/metabolism , Mitochondrial Proteins/metabolism , Peptides/chemistry , Peptides/metabolism , Peptides/therapeutic use
15.
Virus Res ; 130(1-2): 85-95, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17604869

ABSTRACT

Ribonuclease L (RNase L) is an antiviral endoribonuclease that cleaves hepatitis C virus (HCV) RNA at single-stranded UA and UU dinucleotides throughout the open reading frame (ORF). To determine whether RNase L exerts evolutionary pressure on HCV we examined the frequencies of UA and UU dinucleotides in 162 RNA sequences from the Los Alamos National Labs HCV Database (http://hcv.lanl.gov). Considering the base composition of the HCV ORFs, both UA and UU dinucleotides were less frequent than predicted in each of 162 HCV RNAs. UA dinucleotides were significantly less frequent than predicted at each of the three codon positions while UU dinucleotides were less frequent than predicted predominantly at the wobble position of codons. UA and UU dinucleotides were among the least abundant dinucleotides in HCV RNA ORFs. Furthermore, HCV genotype 1 RNAs have a lower frequency of UA and UU dinucleotides than genotype 2 and 3 RNAs, perhaps contributing to increased resistance of HCV genotype 1 infections to interferon therapy. In vitro, RNase L cleaved both HCV genotype 1 and 2 RNAs efficiently. Thus, RNase L can cleave HCV RNAs efficiently and variably reduced frequencies of UA and UU dinucleotides in HCV RNA ORFs are consistent with the selective pressure of RNase L.


Subject(s)
Base Composition/genetics , Dinucleoside Phosphates/genetics , Endoribonucleases/metabolism , Hepacivirus/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , Codon/genetics , Endoribonucleases/immunology , Hepacivirus/immunology , Selection, Genetic
16.
Proc Natl Acad Sci U S A ; 104(23): 9585-90, 2007 Jun 05.
Article in English | MEDLINE | ID: mdl-17535916

ABSTRACT

RNase L, a principal mediator of innate immunity to viral infections in higher vertebrates, is required for a complete IFN antiviral response against certain RNA stranded viruses. dsRNA produced during viral infections activates IFN-inducible synthetases that produce 5'-phosphorylated, 2',5'-oligoadenylates (2-5A) from ATP. 2-5A activates RNase L in a wide range of different mammalian cell types, thus blocking viral replication. However, 2-5A has unfavorable pharmacologic properties; it is rapidly degraded, does not transit cell membranes, and leads to apoptosis. To obtain activators of RNase L with improved drug-like properties, high-throughput screening was performed on chemical libraries by using fluorescence resonance energy transfer. Seven compounds were obtained that activated RNase L at micromolar concentrations, and structure-activity relationship studies resulted in identification of an additional four active compounds. Two lead compounds were shown to have a similar mechanistic path toward RNase L activation as the natural activator 2-5A. The compounds bound to the 2-5A-binding domain of RNase L (as determined by surface plasmon resonance and confirmed by computational docking), and the compounds induced RNase L dimerization and activation. Interestingly, the low-molecular-weight activators of RNase L had broad-spectrum antiviral activity against diverse types of RNA viruses, including the human pathogen human parainfluenza virus type 3, yet these compounds by themselves were not cytotoxic at the effective concentrations. Therefore, these RNase L activators are prototypes for a previously uncharacterized class of broad-spectrum antiviral agents.


Subject(s)
Antiviral Agents/metabolism , Endoribonucleases/metabolism , Enzyme Activators/metabolism , Immunity, Innate/physiology , Parainfluenza Virus 3, Human/metabolism , Adenine Nucleotides/metabolism , Animals , Antiviral Agents/pharmacology , Cell Line , Chromatography, High Pressure Liquid , Dimerization , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Activators/pharmacology , Fluorescence Resonance Energy Transfer , Mice , Models, Molecular , Oligonucleotides/genetics , Oligoribonucleotides/metabolism , Parainfluenza Virus 3, Human/drug effects , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship , Surface Plasmon Resonance , Virus Replication/drug effects
17.
J Virol ; 81(11): 5561-72, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17344297

ABSTRACT

RNase L is an antiviral endoribonuclease that cleaves viral mRNAs after single-stranded UA and UU dinucleotides. Poliovirus (PV) mRNA is surprisingly resistant to cleavage by RNase L due to an RNA structure in the 3C(Pro) open reading frame (ORF). The RNA structure associated with the inhibition of RNase L is phylogenetically conserved in group C enteroviruses, including PV type 1 (PV1), PV2, PV3, coxsackie A virus 11 (CAV11), CAV13, CAV17, CAV20, CAV21, and CAV24. The RNA structure is not present in other human enteroviruses (group A, B, or D enteroviruses). Coxsackievirus B3 mRNA and hepatitis C virus mRNA were fully sensitive to cleavage by RNase L. HeLa cells expressing either wild-type RNase L or a dominant-negative mutant RNase L were used to examine the effects of RNase L on PV replication. PV replication was not inhibited by RNase L activity, but rRNA cleavage characteristic of RNase L activity was detected late during the course of PV infection, after assembly of intracellular virus. Rather than inhibiting PV replication, RNase L activity was associated with larger plaques and better cell-to-cell spread. Mutations in the RNA structure associated with the inhibition of RNase L did not affect the magnitude of PV replication in HeLa cells expressing RNase L, consistent with the absence of observed RNase L activity until after virus assembly. Thus, PV carries an RNA structure in the 3C protease ORF that potently inhibits the endonuclease activity of RNase L, but this RNA structure does not prevent RNase L activity late during the course of infection, as virus assembly nears completion.


Subject(s)
Endoribonucleases/antagonists & inhibitors , Endoribonucleases/chemistry , Open Reading Frames/physiology , Poliovirus/genetics , RNA, Viral/physiology , Amino Acid Sequence , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Base Sequence , Conserved Sequence , Drug Resistance, Viral/genetics , Endoribonucleases/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Poliovirus/physiology
18.
Nucleic Acids Res ; 34(22): 6684-95, 2006.
Article in English | MEDLINE | ID: mdl-17145707

ABSTRACT

The antiviral and antitumor functions of RNase L are enabled by binding to the allosteric effectors 5'-phosphorylated, 2',5'-linked oligoadenylates (2-5A). 2-5A is produced by interferon-inducible 2',5'-oligoadenylate synthetases (OAS) upon activation by viral double-stranded RNA (dsRNA). Because mutations in RNase L have been implicated as risk factors for prostate cancer, we sought to determine if OAS activators are present in prostate cancer cells. We show that prostate cancer cell lines (PC3, LNCaP and DU145), but not normal prostate epithelial cells (PrEC), contain RNA fractions capable of binding to and activating OAS. To identify the RNA activators, we developed a cDNA cloning strategy based on stringent affinity of RNAs for OAS. We thus identified mRNAs for Raf kinase inhibitor protein (RKIP) and poly(rC)-binding protein 2 (PCBP2) that bind and potently activate OAS. In addition, human endogenous retrovirus (hERV) envelope RNAs were present in PC3 cells that bind and activate OAS. Analysis of several gene expression profiling studies indicated that PCBP2 RNA was consistently elevated in metastatic prostate cancer. Results suggest that OAS activation may occur in prostate cancer cells in vivo stimulated by cellular mRNAs for RKIP and PCBP2.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , Phosphatidylethanolamine Binding Protein/genetics , Prostatic Neoplasms/enzymology , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , RNA-Binding Proteins/genetics , Adenine Nucleotides/chemistry , Cell Line, Tumor , Cloning, Molecular , Endogenous Retroviruses/genetics , Enzyme Activation , Gene Products, env/genetics , Humans , Male , Oligoribonucleotides/chemistry , Phosphatidylethanolamine Binding Protein/metabolism , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism
19.
J Biol Chem ; 279(22): 23061-72, 2004 May 28.
Article in English | MEDLINE | ID: mdl-15004022

ABSTRACT

Hyaluronan-binding protein 1 (HABP1) is a trimeric protein with high negative charges distributed asymmetrically along the faces of the molecule. Recently, we have reported that HABP1 exhibits a high degree of structural flexibility, which can be perturbed by ions under in vitro conditions near physiological pH (Jha, B. K., Salunke, D. M., and Datta, K. (2003) J. Biol. Chem. 278, 27464-27472). Here, we report the effect of ionic strength and pH on thermodynamic stability of HABP1. Trimeric HABP1 was shown to unfold reversibly upon dissociation ruling out the possibility of existence of folded monomer. An increase in ionic concentration (0.05-1 M) or decrease in pH (pH 8.0-pH 5.0) induced an unusually high thermodynamic stability of HABP1 as reflected in the gradual increase in transition midpoint temperature, enthalpy of transition, and conformational entropy. Our studies suggest that the presence of counter ions in the molecular environment of HABP1 leads to dramatic reduction of the intramolecular electrostatic repulsion either by de-ionizing the charged amino acid residues or by direct binding leading to a more stable conformation. A regulation on cellular HA-HABP1 interaction by changes in pH and ionic strength may exist, because the more stable conformation attained at higher ionic strength or at acidic pH showed maximum affinity toward HA as probed either in solid phase binding assay on HA-immobilized plates or an in-solution binding assay using intrinsic fluorescence of HABP1.


Subject(s)
Hyaluronan Receptors/chemistry , Carrier Proteins , Cations , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Hydrogen-Ion Concentration , Mitochondrial Proteins , Protein Binding , Protein Conformation , Thermodynamics
20.
J Biol Chem ; 278(30): 27464-72, 2003 Jul 25.
Article in English | MEDLINE | ID: mdl-12719421

ABSTRACT

Hyaluronan-binding protein 1 (HABP1)/p32/gC1qR was characterized as a highly acidic and oligomeric protein, which binds to different ligands like hyaluronan, C1q, and mannosylated albumin. It exists as trimer in high ionic and reducing conditions as shown by crystal structure. In the present study, we have examined the structural changes of HABP1 under a wide range of ionic environments. HABP1 exhibits structural plasticity, which is influenced by the ionic environment under in vitro conditions near physiological pH. At low ionic strength HABP1 exists in a highly expanded and loosely held trimeric structure, similar to that of the molten globule-like state, whereas the presence of salt stabilizes the trimeric structure in a more compact fashion. It is likely that the combination of the high net charge asymmetrically distributed along the faces of the molecule and the relatively low intrinsic hydrophobicity of HABP1 result in its expanded structure at neutral pH. Thus, the addition of counter ions in the molecular environment minimizes the intramolecular electrostatic repulsion in HABP1 leading to its stable and compact conformations, which reflect in its differential binding toward different ligands. Whereas the binding of HABP1 toward HA is enhanced on increasing the ionic strength, no significant effect was observed with the two other ligands, C1q and mannosylated albumin. Thus, although HA interacts only with compact HABP1, C1q and mannosylated albumin can bind to loosely held oligomeric HABP1 as well. In other words, structural changes in HABP1 mediated by changes in the ionic environment are responsible for recognizing different ligands.


Subject(s)
Hyaluronan Receptors/chemistry , Hyaluronan Receptors/physiology , Anilino Naphthalenesulfonates/pharmacology , Animals , Carrier Proteins , Chromatography, Gel , Circular Dichroism , Complement C1q/chemistry , Cross-Linking Reagents/pharmacology , Crystallography, X-Ray , Dimerization , Dose-Response Relationship, Drug , Fluorescent Dyes/pharmacology , Hydrogen-Ion Concentration , Indicators and Reagents/pharmacology , Ions , Ligands , Maleimides/pharmacology , Mitochondrial Proteins , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Salts/pharmacology , Spectrometry, Fluorescence , Static Electricity
SELECTION OF CITATIONS
SEARCH DETAIL
...