Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Nat Mater ; 22(3): 369-379, 2023 03.
Article in English | MEDLINE | ID: mdl-36443576

ABSTRACT

Messenger RNA has now been used to vaccinate millions of people. However, the diversity of pulmonary pathologies, including infections, genetic disorders, asthma and others, reveals the lung as an important organ to directly target for future RNA therapeutics and preventatives. Here we report the screening of 166 polymeric nanoparticle formulations for functional delivery to the lungs, obtained from a combinatorial synthesis approach combined with a low-dead-volume nose-only inhalation system for mice. We identify P76, a poly-ß-amino-thio-ester polymer, that exhibits increased expression over formulations lacking the thiol component, delivery to different animal species with varying RNA cargos and low toxicity. P76 allows for dose sparing when delivering an mRNA-expressed Cas13a-mediated treatment in a SARS-CoV-2 challenge model, resulting in similar efficacy to a 20-fold higher dose of a neutralizing antibody. Overall, the combinatorial synthesis approach allowed for the discovery of promising polymeric formulations for future RNA pharmaceutical development for the lungs.


Subject(s)
COVID-19 , Animals , Mice , RNA, Messenger/genetics , SARS-CoV-2/genetics , Polymers/metabolism , Lung , RNA/metabolism
2.
J Virol ; 96(19): e0134422, 2022 10 12.
Article in English | MEDLINE | ID: mdl-36125302

ABSTRACT

Subtype H7 avian influenza A viruses (IAVs) are enzootic in wild aquatic birds and have caused sporadic spillovers into domestic poultry and humans. Here, we determined the distribution of fucosylated α2,3 sialoglycan (i.e., sialyl Lewis X [SLeX]) in chickens and five common dabbling duck species and the association between SLeX and cell/tissue/host tropisms of H7 IAVs. Receptor binding analyses showed that H7 IAVs bind to both α2,3-linked (SA2,3Gal) and α2,6-linked sialic acids (SA2,6Gal), but with a higher preference for SLeX; H7 IAVs replicated more efficiently in SLeX-overexpressed than SLeX-deficient MDCK cells. While chickens and all tested dabbling ducks expressed abundant SA2,3Gal and SA2,6Gal, SLeX was detected in both respiratory and gastrointestinal tissues of chickens and mallard ducks and in only the respiratory tissues of gadwall, green-wing teal, and northern shoveler but not in wood ducks. Viral-tissue binding assays showed that H7 IAVs bind to chicken colon crypt cells that express SLeX but fewer bind to mallard colon crypt cells, which do not express SLeX; H7 IAVs bind efficiently to epithelial cells of all tissues expressing SA2,3Gal. High viral replication was identified in both chickens and mallards infected with an H7 virus, regardless of SLeX expression, and viruses were detected in all cells to the same degree as viruses detected in the viral-tissue binding assays. In summary, this study suggests that SLeX facilitates infection of H7 viruses, but other types of SA2,3Gal glycan receptors shape the tissue/host tropisms of H7 IAVs. IMPORTANCE In addition to causing outbreaks in domestic poultry, subtype H7 IAVs can cause sporadic spillover infections in lower mammals and humans. In this study, we showed that SLeX expression varies among wild dabbling ducks. Although it facilitated virus binding and affected infection of H7 IAV in cells, SLeX expression is not the only determinant of viral replication at either the tissue or host level. This study suggested that access to heterologous SA2,3Gal glycan receptors, including fucosylated α2,3-linked sialoglycans, shape tissue and host tropism of H7 IAVs in aquatic wild birds.


Subject(s)
Influenza A virus , Influenza in Birds , Sialyl Lewis X Antigen , Viral Tropism , Animals , Animals, Wild/virology , Chickens/virology , Dogs , Ducks/virology , Influenza A virus/pathogenicity , Influenza A virus/physiology , Madin Darby Canine Kidney Cells , Polysaccharides , Sialic Acids , Sialyl Lewis X Antigen/metabolism
3.
Immunity ; 39(2): 311-323, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23954133

ABSTRACT

Nlrp3 inflammasome activation occurs in response to numerous agonists but the specific mechanism by which this takes place remains unclear. All previously evaluated activators of the Nlrp3 inflammasome induce the generation of mitochondrial reactive oxygen species (ROS), suggesting a model in which ROS is a required upstream mediator of Nlrp3 inflammasome activation. Here we have identified the oxazolidinone antibiotic linezolid as a Nlrp3 agonist that activates the Nlrp3 inflammasome independently of ROS. The pathways for ROS-dependent and ROS-independent Nlrp3 activation converged upon mitochondrial dysfunction and specifically the mitochondrial lipid cardiolipin. Cardiolipin bound to Nlrp3 directly and interference with cardiolipin synthesis specifically inhibited Nlrp3 inflammasome activation. Together these data suggest that mitochondria play a critical role in the activation of the Nlrp3 inflammasome through the direct binding of Nlrp3 to cardiolipin.


Subject(s)
Cardiolipins/metabolism , Carrier Proteins/metabolism , Inflammasomes/metabolism , Mitochondria/metabolism , Acetamides/metabolism , Acetamides/pharmacology , Animals , Cardiolipins/immunology , Cell Line , Cyclosporine/metabolism , Enzyme Activation , Humans , Inflammation/chemically induced , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Linezolid , Macrophages/immunology , Macrophages/metabolism , Mice , Mitochondria/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Oxazolidinones/metabolism , Oxazolidinones/pharmacology , Potassium/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/metabolism
4.
J Virol ; 94(24)2020 11 23.
Article in English | MEDLINE | ID: mdl-32967956

ABSTRACT

Wild aquatic birds maintain a large, genetically diverse pool of influenza A viruses (IAVs), which can be transmitted to lower mammals and, ultimately, humans. Through phenotypic analyses of viral replication efficiency, only a small set of avian IAVs were found to replicate well in epithelial cells of the swine upper respiratory tract, and these viruses were shown to infect and cause virus shedding in pigs. Such a phenotypic trait of the viral replication efficiency appears to emerge randomly and is distributed among IAVs across multiple avian species and geographic and temporal orders. It is not determined by receptor binding preference but is determined by other markers across genomic segments, such as those in the ribonucleoprotein complex. This study demonstrates that phenotypic variants of viral replication efficiency exist among avian IAVs but that only a few of these may result in viral shedding in pigs upon infection, providing opportunities for these viruses to become adapted to pigs, thus posing a higher potential risk for creating novel variants or detrimental reassortants within pig populations.IMPORTANCE Swine serve as a mixing vessel for generating pandemic strains of human influenza virus. All hemagglutinin subtypes of IAVs can infect swine; however, only sporadic cases of infection with avian IAVs are reported in domestic swine. The molecular mechanisms affecting the ability of avian IAVs to infect swine are still not fully understood. From the findings of phenotypic analyses, this study suggests that the tissue tropisms (i.e., in swine upper respiratory tracts) of avian IAVs affect their spillovers from wild birds to pigs. It was found that this phenotype is determined not by receptor binding preference but is determined by other markers across genomic segments, such as those in the ribonucleoprotein complex. In addition, our results show that such a phenotypic trait was sporadically and randomly distributed among IAVs across multiple avian species and geographic and temporal orders. This study suggests an efficient way for assessment of the risk posed by avian IAVs, such as in evaluating their potentials to be transmitted from birds to pigs.


Subject(s)
Animals, Wild/virology , Birds/virology , Influenza A virus/genetics , Influenza in Birds/transmission , Influenza in Birds/virology , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Tropism , Animals , Cell Line , Epithelial Cells/virology , HEK293 Cells , Hemagglutinins , Humans , Influenza A virus/growth & development , Pandemics , Phylogeny , Respiratory System/virology , Swine , Virus Replication , Virus Shedding
5.
Mol Ther ; 28(3): 805-819, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31995741

ABSTRACT

There is a clear need for low-cost, self-applied, long-lasting approaches to prevent human immunodeficiency virus (HIV) infection in both men and women, even with the advent of pre-exposure prophylaxis (PrEP). Broadly neutralizing antibodies represent an option to improve HIV prophylaxis, but intravenous delivery, cold-chain stability requirements, low cervicovaginal concentrations, and cost may preclude their use. Here, we present an approach to express the anti-GP120 broadly neutralizing antibody PGT121 in the primary site of inoculation, the female reproductive tract, using synthetic mRNA. Expression is achieved through aerosol delivery of unformulated mRNA in water. We demonstrated high levels of antibody expression for over 28 days with a single mRNA administration in the reproductive tract of sheep. In rhesus macaques, neutralizing antibody titers in secretions developed within 4 h and simian-HIV (SHIV) infection of ex vivo explants was prevented. Persistence of PGT121 in vaginal secretions and epithelium was achieved through the incorporation of a glycosylphosphatidylinositol (GPI) anchor into the heavy chain of the antibody. Overall, we present a new paradigm to deliver neutralizing antibodies to the female reproductive tract for the prevention of HIV infections.


Subject(s)
Broadly Neutralizing Antibodies/immunology , Gene Expression , HIV Antibodies/immunology , Mucous Membrane/immunology , Mucous Membrane/metabolism , RNA, Messenger/administration & dosage , Vagina , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Aerosols , Animals , Chlorocebus aethiops , Female , Fluorescent Antibody Technique , HIV Infections/immunology , HIV-1/immunology , Mice , Neutralization Tests , RNA, Messenger/chemical synthesis , Sheep , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Vagina/immunology , Vagina/metabolism , Vero Cells
6.
J Bacteriol ; 202(10)2020 04 27.
Article in English | MEDLINE | ID: mdl-32179628

ABSTRACT

The capacity of Listeria monocytogenes to adapt to environmental changes is facilitated by a large number of regulatory proteins encoded by its genome. Among these proteins are the uncharacterized LysR-type transcriptional regulators (LTTRs). LTTRs can work as positive and/or negative transcription regulators at both local and global genetic levels. Previously, our group determined by comparative genome analysis that one member of the LTTRs (NCBI accession no. WP_003734782) was present in pathogenic strains but absent from nonpathogenic strains. The goal of the present study was to assess the importance of this transcription factor in the virulence of L. monocytogenes strain F2365 and to identify its regulons. An L. monocytogenes strain lacking lysR (the F2365ΔlysR strain) displayed significant reductions in cell invasion of and adhesion to Caco-2 cells. In plaque assays, the deletion of lysR resulted in a 42.86% decrease in plaque number and a 13.48% decrease in average plaque size. Furthermore, the deletion of lysR also attenuated the virulence of L. monocytogenes in mice following oral and intraperitoneal inoculation. The analysis of transcriptomics revealed that the transcript levels of 139 genes were upregulated, while 113 genes were downregulated in the F2365ΔlysR strain compared to levels in the wild-type bacteria. lysR-repressed genes included ABC transporters, important for starch and sucrose metabolism as well as glycerolipid metabolism, flagellar assembly, quorum sensing, and glycolysis/gluconeogenesis. Conversely, lysR activated the expression of genes related to fructose and mannose metabolism, cationic antimicrobial peptide (CAMP) resistance, and beta-lactam resistance. These data suggested that lysR contributed to L. monocytogenes virulence by broad impact on multiple pathways of gene expression.IMPORTANCEListeria monocytogenes is the causative agent of listeriosis, an infectious and fatal disease of animals and humans. In this study, we have shown that lysR contributes to Listeria pathogenesis and replication in cell lines. We also highlight the importance of lysR in regulating the transcription of genes involved in different pathways that might be essential for the growth and persistence of L. monocytogenes in the host or under nutrient limitation. Better understanding L. monocytogenes pathogenesis and the role of various virulence factors is necessary for further development of prevention and control strategies.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Regulon , Transcription Factors/metabolism , Animals , Bacterial Proteins/genetics , Caco-2 Cells , Female , Gene Expression Regulation, Bacterial , Humans , Listeria monocytogenes/genetics , Mice , Mice, Inbred BALB C , Transcription Factors/genetics , Virulence
7.
J Virol ; 93(13)2019 07 01.
Article in English | MEDLINE | ID: mdl-30996092

ABSTRACT

Subtype H10 influenza A viruses (IAVs) have been recovered from domestic poultry and various aquatic bird species, and sporadic transmission of these IAVs from avian species to mammals (i.e., human, seal, and mink) are well documented. In 2015, we isolated four H10N7 viruses from gulls in Iceland. Genomic analyses showed four gene segments in the viruses were genetically associated with H10 IAVs that caused influenza outbreaks and deaths among European seals in 2014. Antigenic characterization suggested minimal antigenic variation among these H10N7 isolates and other archived H10 viruses recovered from human, seal, mink, and various avian species in Asia, Europe, and North America. Glycan binding preference analyses suggested that, similar to other avian-origin H10 IAVs, these gull-origin H10N7 IAVs bound to both avian-like alpha 2,3-linked sialic acids and human-like alpha 2,6-linked sialic acids. However, when the gull-origin viruses were compared with another Eurasian avian-origin H10N8 IAV, which caused human infections, the gull-origin virus showed significantly higher binding affinity to human-like glycan receptors. Results from a ferret experiment demonstrated that a gull-origin H10N7 IAV replicated well in turbinate, trachea, and lung, but replication was most efficient in turbinate and trachea. This gull-origin H10N7 virus can be transmitted between ferrets through the direct contact and aerosol routes, without prior adaptation. Gulls share their habitat with other birds and mammals and have frequent contact with humans; therefore, gull-origin H10N7 IAVs could pose a risk to public health. Surveillance and monitoring of these IAVs at the wild bird-human interface should be continued.IMPORTANCE Subtype H10 avian influenza A viruses (IAVs) have caused sporadic human infections and enzootic outbreaks among seals. In the fall of 2015, H10N7 viruses were recovered from gulls in Iceland, and genomic analyses showed that the viruses were genetically related with IAVs that caused outbreaks among seals in Europe a year earlier. These gull-origin viruses showed high binding affinity to human-like glycan receptors. Transmission studies in ferrets demonstrated that the gull-origin IAV could infect ferrets, and that the virus could be transmitted between ferrets through direct contact and aerosol droplets. This study demonstrated that avian H10 IAV can infect mammals and be transmitted among them without adaptation. Thus, avian H10 IAV is a candidate for influenza pandemic preparedness and should be monitored in wildlife and at the animal-human interface.


Subject(s)
Ferrets/virology , Influenza A Virus, H10N7 Subtype/pathogenicity , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Aerosols , Animals , Animals, Wild/virology , Birds/virology , Cell Line , Charadriiformes/virology , Genome, Viral , Humans , Iceland , Influenza A Virus, H10N7 Subtype/classification , Influenza A Virus, H10N7 Subtype/genetics , Influenza A Virus, H10N7 Subtype/isolation & purification , Influenza in Birds/virology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/pathology , Pandemics , Phylogeny , Polysaccharides , Respiratory System/pathology , Respiratory System/virology , Sequence Alignment
8.
PLoS Pathog ; 14(12): e1007417, 2018 12.
Article in English | MEDLINE | ID: mdl-30507946

ABSTRACT

Genetic reassortment between influenza A viruses (IAVs) facilitate emergence of pandemic strains, and swine are proposed as a "mixing vessel" for generating reassortants of avian and mammalian IAVs that could be of risk to mammals, including humans. However, how a transmissible reassortant emerges in swine are not well understood. Genomic analyses of 571 isolates recovered from nasal wash samples and respiratory tract tissues of a group of co-housed pigs (influenza-seronegative, avian H1N1 IAV-infected, and swine H3N2 IAV-infected pigs) identified 30 distinct genotypes of reassortants. Viruses recovered from lower respiratory tract tissues had the largest genomic diversity, and those recovered from turbinates and nasal wash fluids had the least. Reassortants from lower respiratory tracts had the largest variations in growth kinetics in respiratory tract epithelial cells, and the cold temperature in swine nasal cells seemed to select the type of reassortant viruses shed by the pigs. One reassortant in nasal wash samples was consistently identified in upper, middle, and lower respiratory tract tissues, and it was confirmed to be transmitted efficiently between pigs. Study findings suggest that, during mixed infections of avian and swine IAVs, genetic reassortments are likely to occur in the lower respiratory track, and tissue tropism is an important factor selecting for a transmissible reassortant.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Orthomyxoviridae Infections , Recombination, Genetic/genetics , Viral Tropism , Animals , Coinfection , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/pathogenicity , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/transmission , Reassortant Viruses/genetics , Reassortant Viruses/pathogenicity , Respiratory Tract Infections/virology , Swine
9.
J Virol ; 92(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29997206

ABSTRACT

An outbreak of respiratory disease caused by the equine-origin influenza A(H3N8) virus was first detected in dogs in 2004 and since then has been enzootic among dogs. Currently, the molecular mechanisms underlying host adaption of this virus from horses to dogs is unknown. Here, we have applied quantitative binding, growth kinetics, and immunofluorescence analyses to elucidate these mechanisms. Our findings suggest that a substitution of W222L in the hemagglutinin of the equine-origin A(H3N8) virus facilitated its host adaption to dogs. This mutation increased binding avidity of the virus specifically to receptor glycans with N-glycolylneuraminic acid (Neu5Gc) and sialyl Lewis X (SLeX) motifs. We have demonstrated these motifs are abundantly located in the submucosal glands of dog trachea. Our findings also suggest that in addition to the type of glycosidic linkage (e.g., α2,3-linkage or α2,6-linkage), the type of sialic acid (Neu5Gc or 5-N-acetyl neuraminic acid) and the glycan substructure (e.g., SLeX) also play an important role in host tropism of influenza A viruses.IMPORTANCE Influenza A viruses (IAVs) cause a significant burden on human and animal health, and mechanisms for interspecies transmission of IAVs are far from being understood. Findings from this study suggest that an equine-origin A(H3N8) IAV with mutation W222L at its hemagglutinin increased binding to canine-specific receptors with sialyl Lewis X and Neu5Gc motifs and, thereby, may have facilitated viral adaption from horses to dogs. These findings suggest that in addition to the glycosidic linkage (e.g., α2,3-linked and α2,6-linked), the substructure in the receptor saccharides (e.g., sialyl Lewis X and Neu5Gc) could present an interspecies transmission barrier for IAVs and drive viral mutations to overcome such barriers.


Subject(s)
Hemagglutinins/genetics , Host Specificity , Influenza A Virus, H3N8 Subtype/genetics , Mutation , Receptors, Virus/genetics , Animals , Binding Sites , Dogs , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Horses , Influenza A Virus, H3N8 Subtype/growth & development , Influenza A Virus, H3N8 Subtype/metabolism , Kinetics , Neuraminic Acids/analysis , Oligosaccharides/analysis , Orthomyxoviridae Infections/virology , Protein Binding , Receptors, Virus/metabolism , Sialyl Lewis X Antigen , Trachea/chemistry , Trachea/virology , Viral Tropism , Virus Attachment
10.
Emerg Infect Dis ; 24(6): 1020-1028, 2018 06.
Article in English | MEDLINE | ID: mdl-29774857

ABSTRACT

Influenza D virus (IDV) has been identified in domestic cattle, swine, camelid, and small ruminant populations across North America, Europe, Asia, South America, and Africa. Our study investigated seroprevalence and transmissibility of IDV in feral swine. During 2012-2013, we evaluated feral swine populations in 4 US states; of 256 swine tested, 57 (19.1%) were IDV seropositive. Among 96 archived influenza A virus-seropositive feral swine samples collected from 16 US states during 2010-2013, 41 (42.7%) were IDV seropositive. Infection studies demonstrated that IDV-inoculated feral swine shed virus 3-5 days postinoculation and seroconverted at 21 days postinoculation; 50% of in-contact naive feral swine shed virus, seroconverted, or both. Immunohistochemical staining showed viral antigen within epithelial cells of the respiratory tract, including trachea, soft palate, and lungs. Our findings suggest that feral swine might serve an important role in the ecology of IDV.


Subject(s)
Orthomyxoviridae Infections/veterinary , Swine Diseases/epidemiology , Swine Diseases/virology , Thogotovirus , Animals , Female , Genotype , Geography, Medical , Hemagglutination , Hemagglutination Tests , Public Health Surveillance , Seroepidemiologic Studies , Swine , Swine Diseases/diagnosis , Thogotovirus/classification , Thogotovirus/genetics , Thogotovirus/immunology , United States/epidemiology , Viral Load , Virus Shedding , Zoonoses
11.
J Cell Sci ; 129(7): 1455-67, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26872784

ABSTRACT

The role of TRAF2 and TRAF5 in TNFα-induced NF-κB activation has become complicated owing to the accumulation of conflicting data. Here, we report that 7-day-old TRAF2-knockout (KO) and TRAF2 TRAF5 double KO (TRAF2/5-DKO) mice exhibit enhanced canonical IκB kinase (IKK) and caspase-8 activation in spleen and liver, and that subsequent knockout of TNFα suppresses the basal activity of caspase-8, but not of IKK. In primary TRAF2 KO and TRAF2/5-DKO cells, TNFα-induced immediate IKK activation is impaired, whereas delayed IKK activation occurs normally; as such, owing to elevated basal and TNFα-induced delayed IKK activation, TNFα stimulation leads to significantly increased induction of a subset of NF-κB-dependent genes in these cells. In line with this, both TRAF2 KO and TRAF2/5-DKO mice succumb to a sublethal dose of TNFα owing to increased expression of NF-κB target genes, diarrhea and bradypnea. Notably, depletion of IAP1 and IAP2 (also known as BIRC2 and BIRC3, respectively) also results in elevated basal IKK activation that is independent of autocrine TNFα production and that impairs TNFα-induced immediate IKK activation. These data reveal that TRAF2, IAP1 and IAP2, but not TRAF5, cooperatively regulate basal and TNFα-induced immediate IKK activation.


Subject(s)
Caspase 8/metabolism , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 5/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/genetics , Baculoviral IAP Repeat-Containing 3 Protein , Cells, Cultured , Chemokine CCL5/metabolism , Enzyme Activation/genetics , I-kappa B Kinase/genetics , Inhibitor of Apoptosis Proteins/deficiency , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 5/genetics , Tumor Necrosis Factor-alpha/genetics , Ubiquitin-Protein Ligases/deficiency
12.
Mol Cell ; 40(6): 893-904, 2010 Dec 22.
Article in English | MEDLINE | ID: mdl-21172655

ABSTRACT

Genetic deletion of the mitochondrial deacetylase sirtuin-3 (Sirt3) results in increased mitochondrial superoxide, a tumor-permissive environment, and mammary tumor development. MnSOD contains a nutrient- and ionizing radiation (IR)-dependent reversible acetyl-lysine that is hyperacetylated in Sirt3⁻/⁻ livers at 3 months of age. Livers of Sirt3⁻/⁻ mice exhibit decreased MnSOD activity, but not immunoreactive protein, relative to wild-type livers. Reintroduction of wild-type but not deacetylation null Sirt3 into Sirt3⁻/⁻ MEFs deacetylated lysine and restored MnSOD activity. Site-directed mutagenesis of MnSOD lysine 122 to an arginine, mimicking deacetylation (lenti-MnSOD(K122-R)), increased MnSOD activity when expressed in MnSOD⁻/⁻ MEFs, suggesting acetylation directly regulates function. Furthermore, infection of Sirt3⁻/⁻ MEFs with lenti-MnSOD(K122-R) inhibited in vitro immortalization by an oncogene (Ras), inhibited IR-induced genomic instability, and decreased mitochondrial superoxide. Finally, IR was unable to induce MnSOD deacetylation or activity in Sirt3⁻/⁻ livers, and these irradiated livers displayed significant IR-induced cell damage and microvacuolization in their hepatocytes.


Subject(s)
Conserved Sequence , Evolution, Molecular , Lysine/metabolism , Oxidative Stress , Sirtuin 3/metabolism , Superoxide Dismutase/metabolism , Acetylation , Animals , Arginine/metabolism , Cell Line , Mice , Mutagenesis, Site-Directed , Sirtuin 3/deficiency , Sirtuin 3/genetics
13.
J Virol ; 90(12): 5636-5642, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27030270

ABSTRACT

UNLABELLED: Cattle have been proposed as the natural reservoir of a novel member of the virus family Orthomyxoviridae, which has been tentatively classified as influenza D virus (IDV). Although isolated from sick animals, it is unclear whether IDV causes any clinical disease in cattle. To address this aspect of Koch's postulates, three dairy calves (treatment animals) held in individual pens were inoculated intranasally with IDV strain D/bovine/Mississippi/C00046N/2014. At 1 day postinoculation, a seronegative calf (contact animal) was added to each of the treatment animal pens. The cattle in both treatment and contact groups seroconverted, and virus was detected in their respiratory tracts. Histologically, there was a significant increase in neutrophil tracking in tracheal epithelia of the treatment calves compared to control animals. While infected and contact animals demonstrated various symptoms of respiratory tract infection, they were mild, and the calves in the treatment group did not differ from the controls in terms of heart rate, respiratory rate, or rectal temperature. To mimic zoonotic transmission, two ferrets were exposed to a plastic toy fomite soaked with infected nasal discharge from the treatment calves. These ferrets did not shed the virus or seroconvert. In summary, this study demonstrates that IDV causes a mild respiratory disease upon experimental infection of cattle and can be transmitted effectively among cattle by in-pen contact, but not from cattle to ferrets through fomite exposure. These findings support the hypothesis that cattle are a natural reservoir for the virus. IMPORTANCE: A novel influenza virus, tentatively classified as influenza D virus (IDV), was identified in swine, cattle, sheep, and goats. Among these hosts, cattle have been proposed as the natural reservoir. In this study, we show that cattle experimentally infected with IDV can shed virus and transmit it to other cattle through direct contact, but not to ferrets through fomite routes. IDV caused minor clinical signs in the infected cattle, fulfilling another of Koch's postulates for this novel agent, although other objective clinical endpoints were not different from those of control animals. Although the disease observed was mild, IDV induced neutrophil tracking and epithelial attenuation in cattle trachea, which could facilitate coinfection with other pathogens, and in doing so, predispose animals to bovine respiratory disease.


Subject(s)
Cattle Diseases/virology , Disease Reservoirs/virology , Orthomyxoviridae Infections/veterinary , Respiratory Tract Infections/veterinary , Thogotovirus/pathogenicity , Animals , Cattle , Ferrets , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Respiratory System/virology , Respiratory Tract Infections/virology , Seroconversion , Thogotovirus/isolation & purification , Trachea/cytology , Trachea/pathology , Trachea/virology , Virus Shedding
14.
Proc Natl Acad Sci U S A ; 111(3): 1072-7, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24395802

ABSTRACT

Chronic recurrent multifocal osteomyelitis (CRMO) is a human autoinflammatory disorder that primarily affects bone. Missense mutation (L98P) of proline-serine-threonine phosphatase-interacting protein 2 (Pstpip2) in mice leads to a disease that is phenotypically similar to CRMO called chronic multifocal osteomyelitis (cmo). Here we show that deficiency of IL-1RI in cmo mice resulted in a significant reduction in the time to onset of disease as well as the degree of bone pathology. Additionally, the proinflammatory cytokine IL-1ß, but not IL-1α, played a critical role in the pathology observed in cmo mice. In contrast, disease in cmo mice was found to be independent of the nucleotide-binding domain, leucine-rich repeat-containing family, pyrin domain-containing 3 (NLRP3) inflammasome as well as caspase-1. Neutrophils, but not bone marrow-derived macrophages, from cmo mice secreted increased IL-1ß in response to ATP, silica, and Pseudomonas aeruginosa compared with neutrophils from WT mice. This aberrant neutrophil response was sensitive to inhibition by serine protease inhibitors. These results demonstrate an inflammasome-independent role for IL-1ß in disease progression of cmo and implicate neutrophils and neutrophil serine proteases in disease pathogenesis. These data provide a rationale for directly targeting IL-1RI or IL-1ß as a therapeutic strategy in CRMO.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cytoskeletal Proteins/genetics , Gene Expression Regulation , Interleukin-1beta/metabolism , Osteomyelitis/immunology , Animals , Bone Marrow Cells/cytology , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Inflammasomes/metabolism , Macrophages/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Mutation, Missense , Neutrophils/cytology , Neutrophils/metabolism , Osteomyelitis/genetics , Protein Structure, Tertiary , Receptors, Interleukin-1/genetics
15.
Hum Mol Genet ; 23(10): 2665-77, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24381307

ABSTRACT

Exome sequencing indicated that the gene encoding the calpain-5 protease, CAPN5, is the likely cause of retinal degeneration and autoimmune uveitis in human patients with autosomal dominant neovascular inflammatory vitreoretinopathy (ADNIV, OMIM #193235). To explore the mechanism of ADNIV, a human CAPN5 disease allele was expressed in mouse retinas with a lentiviral vector created to express either the wild-type human (h) CAPN5 or the ADNIV mutant hCAPN5-R243L allele under a rhodopsin promoter with tandem green fluorescent protein (GFP) expression. Vectors were injected into the subretinal space of perinatal mice. Mouse phenotypes were analyzed using electroretinography, histology and inflammatory gene expression profiling. Mouse calpain-5 showed high homology to its human ortholog with >98% sequence identity that includes the ADNIV mutant residue. Calpain-5 protein was expressed in the inner and outer segments of the photoreceptors and in the outer plexiform layer. Expression of the hCAPN5-R243L allele caused loss of the electroretinogram b-wave, photoreceptor degeneration and induction of immune cell infiltration and inflammatory genes in the retina, recapitulating major features of the ADNIV phenotype. Intraocular neovascularization and fibrosis were not observed during the study period. Our study shows that expression of the hCAPN5-R243L disease allele elicits an ADNIV-like disease in mice. It further suggests that ADNIV is due to CAPN5 gain-of-function rather than haploinsufficiency, and retinal expression may be sufficient to generate an autoimmune response. Genetic models of ADNIV in the mouse can be used to explore protease mechanisms in retinal degeneration and inflammation as well as preclinical therapeutic testing.


Subject(s)
Calpain/genetics , Retina/metabolism , Vitreoretinopathy, Proliferative/genetics , Adult , Amino Acid Sequence , Animals , Calpain/metabolism , Disease Models, Animal , Exome , Humans , Inflammation Mediators/metabolism , Lentivirus/genetics , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Mutation, Missense , Photoreceptor Cells, Vertebrate/pathology , Retina/pathology , Transduction, Genetic , Vitreoretinopathy, Proliferative/immunology , Vitreoretinopathy, Proliferative/pathology
16.
J Immunol ; 193(10): 5190-8, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25320279

ABSTRACT

IgG immune complexes have been shown to modify immune responses driven by APCs in either a pro- or anti-inflammatory direction depending upon the context of stimulation. However, the ability of immune complexes to modulate the inflammasome-dependent innate immune response is unknown. In this study, we show that IgG immune complexes suppress IL-1α and IL-1ß secretion through inhibition of inflammasome activation. The mechanism by which this inhibition occurs is via immune complex ligation of activating FcγRs, resulting in prevention of both activation and assembly of the inflammasome complex in response to nucleotide-binding domain leucine-rich repeat (NLR) P3, NLRC4, or AIM2 agonists. In vivo, administration of Ag in the form of an immune complex during priming of the immune response inhibited resultant adaptive immune responses in an NLRP3-dependent model of allergic airway disease. Our data reveal an unexpected mechanism regulating CD4(+) T cell differentiation, by which immune complexes suppress inflammasome activation and the generation of IL-1α and IL-1ß from APCs, which are critical for the Ag-driven differentiation of CD4(+) T cells.


Subject(s)
Antigen-Antibody Complex/genetics , CD4-Positive T-Lymphocytes/immunology , Inflammasomes/immunology , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Lung/immunology , Respiratory Hypersensitivity/immunology , Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Animals , CD4-Positive T-Lymphocytes/pathology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/pathology , Gene Expression Regulation , Immunity, Innate , Inflammasomes/genetics , Interleukin-1alpha/biosynthesis , Interleukin-1beta/biosynthesis , Lung/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin , Receptors, IgG/genetics , Receptors, IgG/immunology , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/pathology , Signal Transduction
17.
Cell Biochem Funct ; 34(8): 628-639, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27935134

ABSTRACT

Nonalcoholic steatohepatitis is a common liver disease that is often accompanied by dysregulated iron metabolism. The aim of the study was to test the hypothesis that aberrant iron metabolism in nonalcoholic steatohepatitis is modulated by genetic susceptibility to inflammation and oxidative stress. Hepatic histology and iron content were assessed in 3 inbred strains of mice (C57BL/6, BALB/c, and C3H/HeJ) fed an atherogenic diet (AD). Hepatic expression of genes relevant to iron metabolism, inflammation, and oxidative stress were quantitated by real-time reverse transcription-polymerase chain reaction. At 6 weeks on the AD, histologic injury and induction of inflammatory and oxidative stress-associated gene expression were most pronounced in C57BL/6. At 18 weeks on the AD, these parameters were similar in C57BL/6 and BALB/c. Atherogenic diet-fed C3H/HeJ showed milder responses at both time points. The AD was associated with decreased hepatic iron concentrations in all strains at 6 and 18 weeks. The decrease in hepatic iron concentrations did not correlate with changes in hepcidin expression and was not associated with altered expression of iron transporters. These findings are similar to those observed in models of obesity-induced steatosis and indicate that hepatic steatosis can be associated with depletion of iron stores that is not explained by upregulation of hepcidin expression by inflammation. SIGNIFICANCE OF THE STUDY: Nonalcoholic steatohepatitis (NASH) is a common liver disease that often accompanies the metabolic syndrome. The latter condition has been linked to iron deficiency and diminished intestinal iron absorption, likely the result of hepcidin upregulation by chronic inflammation. Paradoxically, some NASH patients accumulate excess hepatic iron, which may increase fibrosis and cancer risk. Iron accumulation has been attributed to suppression of hepcidin by oxidative stress. The objective of this study was to investigate the contributions of inflammation and oxidative stress to altered hepatic iron metabolism in a murine model of NASH using inbred strains of mice with differing susceptibilities to injury.


Subject(s)
Iron/metabolism , Liver/metabolism , Liver/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Body Weight , Diet, Atherogenic , Disease Models, Animal , Female , Gene Expression Regulation , Hepcidins/metabolism , Inflammation/genetics , Inflammation/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/genetics , Organ Size , Oxidative Stress/genetics , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Statistics, Nonparametric , Time Factors
18.
Am J Physiol Gastrointest Liver Physiol ; 308(6): G459-71, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25591863

ABSTRACT

Multiple organ systems, including the gastrointestinal tract, pancreas, and hepatobiliary systems, are affected by cystic fibrosis (CF). Many of these changes begin early in life and are difficult to study in young CF patients. Recent development of novel CF animal models has expanded opportunities in the field to better understand CF pathogenesis and evaluate traditional and innovative therapeutics. In this review, we discuss manifestations of CF disease in gastrointestinal, pancreatic, and hepatobiliary systems of humans and animal models. We also compare the similarities and limitations of animal models and discuss future directions for modeling CF.


Subject(s)
Biliary Tract Diseases/physiopathology , Cystic Fibrosis/complications , Gastrointestinal Diseases/physiopathology , Liver Diseases/physiopathology , Pancreatic Diseases/physiopathology , Animals , Biliary Tract Diseases/etiology , Biliary Tract Diseases/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Disease Models, Animal , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/metabolism , Genetic Predisposition to Disease , Humans , Liver Diseases/etiology , Liver Diseases/metabolism , Mice, Inbred CFTR , Mutation , Pancreatic Diseases/etiology , Pancreatic Diseases/metabolism , Phenotype , Species Specificity
19.
Am J Pathol ; 184(5): 1309-22, 2014 May.
Article in English | MEDLINE | ID: mdl-24637292

ABSTRACT

Cystic fibrosis (CF) is a multiorgan disease caused by loss of a functional cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel in many epithelia of the body. Here we report the pathology observed in the gastrointestinal organs of juvenile to adult CFTR-knockout ferrets. CF gastrointestinal manifestations included gastric ulceration, intestinal bacterial overgrowth with villous atrophy, and rectal prolapse. Metagenomic phylogenetic analysis of fecal microbiota by deep sequencing revealed considerable genotype-independent microbial diversity between animals, with the majority of taxa overlapping between CF and non-CF pairs. CF hepatic manifestations were variable, but included steatosis, necrosis, biliary hyperplasia, and biliary fibrosis. Gallbladder cystic mucosal hyperplasia was commonly found in 67% of CF animals. The majority of CF animals (85%) had pancreatic abnormalities, including extensive fibrosis, loss of exocrine pancreas, and islet disorganization. Interestingly, 2 of 13 CF animals retained predominantly normal pancreatic histology (84% to 94%) at time of death. Fecal elastase-1 levels from these CF animals were similar to non-CF controls, whereas all other CF animals evaluated were pancreatic insufficient (<2 µg elastase-1 per gram of feces). These findings suggest that genetic factors likely influence the extent of exocrine pancreas disease in CF ferrets and have implications for the etiology of pancreatic sufficiency in CF patients. In summary, these studies demonstrate that the CF ferret model develops gastrointestinal pathology similar to CF patients.


Subject(s)
Aging/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Gastrointestinal Tract/pathology , Gene Knockout Techniques , Animals , Atrophy , Bacteria/growth & development , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Ferrets , Gastrointestinal Tract/abnormalities , Humans , Mucus/metabolism , Organ Specificity
20.
Ann Surg Oncol ; 22(3): 866-73, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25326397

ABSTRACT

BACKGROUND: Locally advanced breast cancer (LABC) poses complex management issues due to failure of response to chemotherapy and progression to local complications such as skin erosion, superinfection, and lymphedema. Most cell line and animal models are not adequate to study LABC. METHODS: A patient-derived xenograft (IOWA-1T) from a patient with LABC was characterized for expression profile, short tandem repeat profile, oncogenic mutations, xenograft growth, and response to therapy. RESULTS: Short tandem repeat profile authenticated the cell line as derived from a human woman. The primary tumor and derived xenografts were weakly estrogen receptor alpha positive (<5%), progesterone receptor negative, and HER2 nonamplified. Expression array profile compared to MCF-7 and BT-549 cell lines indicate that IOWA-1T was more closely related to basal breast cancer. IOWA-1T harbors a homozygous R248Q mutation of the TP53 gene; in vitro invasion assay was comparable to BT-549 and greater than MCF-7. IOWA-1T xenografts developed palpable tumors in 9.6 ± 1.6 days, compared to 49 ± 13 days for parallel experiments with BT-20 cells (p < 0.002). Tumor xenografts became locally advanced, growing to >2 cm in 21.6 ± 2 days, characterized by skin erosion necessitating euthanasia. The SUMO inhibitor anacardic acid inhibited the outgrowth of IOWA-1T xenografts, while doxorubicin had no effect on tumorigenesis. CONCLUSIONS: IOWA-1T is a novel cell line with an expression pattern consistent with basal breast cancer. Xenografts recapitulated LABC and provide a novel model for testing therapeutic drugs that may be effective in cases resistant to conventional chemotherapy.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Disease Models, Animal , Gene Expression Profiling , Animals , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Cell Proliferation , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL