Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters










Publication year range
1.
Clin Exp Allergy ; 48(6): 691-702, 2018 06.
Article in English | MEDLINE | ID: mdl-29473965

ABSTRACT

BACKGROUND: Eosinophils in the nasal mucosa are an elemental feature of allergic rhinitis. OBJECTIVE: Our objective was to explore eosinophilic inflammation and its impact on respiratory virus infection at the nasal mucosa. METHODS: Inflammation in the nasal mucosae of mice was evaluated in response to repetitive stimulation with strict intranasal volumes of a filtrate of Alternaria alternata. Mice were then challenged with influenza virus. RESULTS: Repetitive stimulation with A. alternata resulted in eosinophil recruitment to the nasal passages in association with elevated levels of IL-5, IL-13 and eotaxin-1; eosinophil recruitment was diminished in eotaxin-1-/- mice, and abolished in Rag1-/- mice. A. alternata also resulted in elevated levels of nasal wash IgA in both wild-type and eosinophil-deficient ∆dblGATA mice. Interestingly, A. alternata-treated mice responded to an influenza virus infection with profound weight loss and mortality compared to mice that received diluent alone (0% vs 100% survival, ***P < .001); the lethal response was blunted when A. alternata was heat-inactivated. Minimal differences in virus titre were detected, and eosinophils present in the nasal passages at the time of virus inoculation provided no protection against the lethal sequelae. Interestingly, nasal wash fluids from mice treated with A. alternata included more neutrophils and higher levels of pro-inflammatory mediators in response to virus challenge, among these, IL-6, a biomarker for disease severity in human influenza. CONCLUSIONS AND CLINICAL RELEVANCE: Repetitive administration of A. alternata resulted in inflammation of the nasal mucosae and unanticipated morbidity and mortality in response to subsequent challenge with influenza virus. Interestingly, and in contrast to findings in the lower airways, eosinophils recruited to the nasal passages provided no protection against lethal infection. As increased susceptibility to influenza virus among individuals with rhinitis has been the subject of several clinical reports, this model may be used for further exploration of these observations.


Subject(s)
Alternaria/immunology , Eosinophilia/immunology , Eosinophils/immunology , Influenza A virus/immunology , Nasal Mucosa/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Animals , Biomarkers , Coinfection , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Eosinophilia/pathology , Female , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunization , Inflammation Mediators/metabolism , Mice , Mice, Knockout , Nasal Mucosa/metabolism , Nasal Mucosa/microbiology , Nasal Mucosa/virology , Neutrophil Infiltration/immunology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology
2.
Clin Exp Allergy ; 48(1): 29-38, 2018 01.
Article in English | MEDLINE | ID: mdl-29068539

ABSTRACT

BACKGROUND: Eosinophils are immunomodulatory leucocytes that contribute to the pathogenesis of Th2-driven asthma and allergic lung diseases. OBJECTIVE: Our goal was to identify unique properties of eosinophils recruited to the lungs and airways of mice in response to challenge with asthma-associated fungal allergens. METHODS: Mice were challenged intranasally on days 0, 3 and 6 with a filtrate of Alternaria alternata. Recruited eosinophils were enumerated in bronchoalveolar lavage fluid. Eosinophils were also isolated from lungs of mice sensitized and challenged with Aspergillus fumigatus and evaluated ex vivo in tissue culture. RESULTS: Eosinophils persist in the airways for several weeks in response to brief provocation with A. alternata in wild-type, Gm-csf- and eotaxin-1-gene-deleted mice, while eosinophils are recruited but do not persist in the absence of IL-13. Eosinophils isolated from the lungs A. alternata-challenged mice are cytokine-enriched compared to those from IL5tg mice, including 800-fold higher levels of eotaxin-1. Furthermore, eosinophils from the lungs and spleen of fungal allergen-challenged wild-type mice are capable of prolonged survival ex vivo, in contrast to eosinophils from both untreated and fungal allergen-challenged IL5tg mice, which undergo rapid demise in the absence of exogenous cytokine support. TNF-α (but not IL5, IL-3, eotaxin-1 or GM-CSF) was detected in supernatants of ex vivo eosinophil cultures from the lungs of fungal allergen-challenged wild-type mice. However, neither TNF-α gene deletion nor anti-TNF-α neutralizing antibodies had any impact sustained eosinophil survival ex vivo. CONCLUSION AND CLINICAL RELEVANCE: Eosinophils are phenotypically and functionally heterogeneous. As shown here, eosinophils from fungal allergen-challenged wild-type mice maintain a distinct cytokine profile, and, unlike eosinophils isolated from IL5tg mice, they survive ex vivo in the absence of exogenous pro-survival cytokine support. As treatments for asthma currently in development focus on limiting eosinophil viability via strategic cytokine blockade, the molecular mechanisms underlying differential survival merit further investigation.


Subject(s)
Allergens/toxicity , Alternaria/immunology , Antigens, Fungal/toxicity , Aspergillus fumigatus/immunology , Asthma/immunology , Eosinophils/immunology , Allergens/immunology , Animals , Antigens, Fungal/immunology , Asthma/chemically induced , Asthma/pathology , Eosinophils/pathology , Female , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Th2 Cells/immunology , Th2 Cells/pathology
4.
Curr Med Chem ; 19(10): 1424-31, 2012.
Article in English | MEDLINE | ID: mdl-22360479

ABSTRACT

Respiratory syncytial virus (RSV; Family Paramyxoviridae, Genus Pneumovirus) is a major respiratory pathogen of infants and children and an emerging pathogen of the elderly. Current management of RSV disease includes monoclonal antibody prophylaxis for infants identified as high risk and supportive care for those with active infection; there is no vaccine, although several are under study. In this manuscript, we review published findings from human autopsy studies, as well as experiments that focus on human clinical samples and mouse models of acute pneumovirus infection that elucidate basic principles of disease pathogenesis. Consideration of these data suggests that the inflammatory responses to RSV and related pneumoviral pathogens can be strong, persistent, and beyond the control of conventional antiviral and anti-inflammatory therapies, and can have profound negative consequences to the host. From this perspective, we consider the case for specific immunomodulatory strategies that may have the potential to alleviate some of the more serious sequelae of this disease.


Subject(s)
Immunologic Factors/therapeutic use , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antiviral Agents/therapeutic use , Cytokines/antagonists & inhibitors , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Humans , Oligodeoxyribonucleotides, Antisense/therapeutic use , Pneumovirus/genetics , Pneumovirus/immunology , Pneumovirus Infections/drug therapy , Pneumovirus Infections/immunology , Pneumovirus Infections/pathology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus, Human/genetics
6.
Curr Pharm Biotechnol ; 9(3): 135-40, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18673278

ABSTRACT

The eosinophil-derived neurotoxin (EDN, also known as eosinophil protein-X) is best-known as one of the four major proteins found in the large specific granules of human eosinophilic leukocytes. Although it was named for its discovery and initial characterization as a neurotoxin, it is also expressed constitutively in human liver tissue and its expression can be induced in macrophages by proinflammatory stimuli. EDN and its divergent orthologs in rodents have ribonuclease activity, and are members of the extensive RNase A superfamily, although the relationship between the characterized physiologic functions and enzymatic activity remains poorly understood. Recent explorations into potential physiologic functions for EDN have provided us with some insights into its role in antiviral host defense, as a chemoattractant for human dendritic cells, and most recently, as an endogenous ligand for toll-like receptor (TLR)2.


Subject(s)
Eosinophil-Derived Neurotoxin , Eosinophils/enzymology , Animals , Dendritic Cells/drug effects , Eosinophil-Derived Neurotoxin/chemistry , Eosinophil-Derived Neurotoxin/genetics , Eosinophil-Derived Neurotoxin/physiology , Eosinophil-Derived Neurotoxin/toxicity , Humans , Ligands , Models, Molecular , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Phylogeny , Toll-Like Receptor 2/metabolism
7.
Genes Immun ; 5(8): 668-74, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15526002

ABSTRACT

The eosinophil-associated ribonucleases (Ears) are rapidly evolving proteins found in multigene clusters that are unique to each rodent species. Of the 15 independent genes in the Mus musculus cluster, only mEars 1 and 2 are expressed at significant levels at homeostasis. Here we characterize the expression of mEar 6 in the liver and spleen in mice in response to infection with the helminthic parasite, Schistosoma mansoni. Interestingly, expression of mEar 6 is not directly related to the elevated levels of serum IL-5 or tissue eosinophilia characteristic of this disease, as no mEar 6 transcripts were detected in the liver or the spleen from uninfected IL-5-transgenic mice. The coding sequence of mEar 6 has diverged under positive selection pressure (K(a)/K(s) > 1.0) and has a unique unpaired cysteine near the carboxy-terminus of the protein. The high catalytic efficiency of recombinant mEar 6 (k(cat)/K(m) = 0.9 x 10(6)/M/s) is similar to that of the cluster's closest human ortholog, eosinophil-derived neurotoxin (EDN/RNase 2). In summary, we have identified mEar 6 as one of only two RNase A superfamily ribonucleases known to be expressed specifically in response to pathophysiologic stress in vivo.


Subject(s)
Eosinophil Cationic Protein/genetics , Gene Expression , Schistosoma mansoni , Schistosomiasis mansoni/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , DNA Primers , Eosinophil Cationic Protein/metabolism , Evolution, Molecular , Immunoblotting , Liver/metabolism , Liver/pathology , Mice , Molecular Sequence Data , Recombinant Proteins/metabolism , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/pathology , Selection, Genetic , Sequence Alignment , Sequence Analysis, DNA , Spleen/metabolism , Spleen/pathology
8.
J Infect Dis ; 184(12): 1518-23, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11740726

ABSTRACT

The use of glucocorticoids for the treatment of symptoms associated with respiratory syncytial virus (RSV) infection has been questioned. To evaluate the sequelae of glucocorticoid administration in the setting of pneumovirus infection in vivo, hydrocortisone was administered to mice infected with pneumonia virus of mice (PVM), a pneumovirus and natural rodent pathogen that is closely related to RSV and replicates the signs and symptoms of severe human RSV infection. Results showed that hydrocortisone spared the pulmonary neutrophilia but resulted in ablation of the pulmonary eosinophilia, despite continued production of the relevant chemoattractant, macrophage inflammatory protein-1alpha. Hydrocortisone also led to diminished production of inducible nitric oxide synthase and accumulation of reactive nitrogen species in lung tissue and bronchoalveolar lavage fluid and diminished lymphocyte recruitment. PVM-infected mice responded to hydrocortisone with enhanced viral replication and accelerated mortality. These results suggest several mechanisms to explain why glucocorticoid therapy may be of limited benefit in the overall picture of pneumovirus infection.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Hydrocortisone/administration & dosage , Murine pneumonia virus/physiology , Pneumonia, Viral/immunology , Pneumovirus Infections/immunology , Animals , Chemokine CCL2/metabolism , Chemokine CCL4 , Disease Models, Animal , Humans , Lung/immunology , Lung/virology , Macrophage Inflammatory Proteins/metabolism , Mice , Mice, Inbred BALB C , Murine pneumonia virus/isolation & purification , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology , Pneumovirus Infections/drug therapy , Pneumovirus Infections/mortality , Pneumovirus Infections/virology , Treatment Outcome , Virus Replication/drug effects
9.
J Leukoc Biol ; 70(5): 691-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11698487

ABSTRACT

Eosinophils remain among the most enigmatic of cells, as our appreciation of their detrimental activities--e.g., asthma and allergic disease--far outweighs our understanding of their beneficial effects. Among the major secretory effector proteins of eosinophils are the ribonucleases eosinophil-derived neurotoxin (EDN) and eosinophil cationic protein (ECP) in primates and their orthologs, the eosinophil-associated ribonucleases (EARs) in rodents. The rapid diversification observed among these ribonucleases suggested that the ultimate target(s) might be similarly efficient at generating sequence diversity while maintaining an unalterable susceptibility to ribonucleolytic cleavage. This has prompted us to consider a role for these proteins and by extension, for eosinophils, in host defense against single-stranded RNA virus pathogens. We detail our studies of the antiviral activity of eosinophils and eosinophil ribonucleases against respiratory syncytial virus (RSV) in vitro and the related, natural rodent pathogen, pneumonia virus of mice (PVM), in vivo, and consider the possibility that antiviral host defense and the dysregulated responses leading to asthma represent opposing sides of an eosinophil-mediated double-edged sword.


Subject(s)
Blood Proteins/physiology , Eosinophils/physiology , Respiratory Tract Infections/immunology , Ribonucleases/physiology , Virus Diseases/immunology , Animals , Asthma/etiology , Asthma/immunology , Chemotaxis , Child , Cytoplasmic Granules/enzymology , Disease Susceptibility , Eosinophil Granule Proteins , Eosinophil-Derived Neurotoxin , Eosinophils/enzymology , Evolution, Molecular , Humans , Infant , Mice , Murine pneumonia virus/immunology , Pneumovirus Infections/immunology , Pneumovirus Infections/virology , Primates , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/virology , Recombinant Proteins/pharmacology , Respiratory Syncytial Virus Infections/immunology , Respiratory Tract Infections/virology , Ribonucleases/genetics , Ribonucleases/pharmacology , Rodentia , Species Specificity
10.
Respir Res ; 2(4): 225-33, 2001.
Article in English | MEDLINE | ID: mdl-11686888

ABSTRACT

Respiratory syncytial virus (RSV) and pneumonia virus of mice (PVM) are viruses of the family Paramyxoviridae, subfamily pneumovirus, which cause clinically important respiratory infections in humans and rodents, respectively. The respiratory epithelial target cells respond to viral infection with specific alterations in gene expression, including production of chemoattractant cytokines, adhesion molecules, elements that are related to the apoptosis response, and others that remain incompletely understood. Here we review our current understanding of these mucosal responses and discuss several genomic approaches, including differential display reverse transcription-polymerase chain reaction (PCR) and gene array strategies, that will permit us to unravel the nature of these responses in a more complete and systematic manner.


Subject(s)
Gene Expression , Murine pneumonia virus , Pneumovirus Infections/genetics , Respiratory Syncytial Virus Infections/genetics , Animals , Apoptosis , Chemokines/biosynthesis , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Oligonucleotide Array Sequence Analysis
12.
J Mol Evol ; 53(1): 31-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11683320

ABSTRACT

We present sequences of five novel RNase A superfamily ribonuclease genes of the bullfrog, Rana catesbeiana. All five genes encode ribonucleases that are similar to Onconase, a cytotoxic ribonuclease isolated from oocytes of R. pipiens. With amino acid sequence data from 14 ribonucleases from three Rana species (R. catesbeiana, R. japonica, and R. pipiens), we have constructed bootstrap-supported phylogenetic trees that reorganize these ribonucleases into five distinct lineages--the pancreatic ribonucleases (RNases 1), the eosinophil-associated ribonucleases (RNases 2, 3, and 6), the ribonucleases 4, the angiogenins (RNases 5) and the Rana ribonucleases--with the Rana ribonucleases no more closely related to the angiogenins than they are to any of the other ribonuclease lineages shown. Further phylogenetic analysis suggests the division of the Rana ribonucleases into two subclusters (A and B), with positive (Darwinian) selection (dN/dS > 1.0) and an elevated rate of radical nonsynonymous substitution (dR) contributing to the rapid diversification of ribonucleases within each cluster. This pattern of evolution-rapid diversification via positive selection among sequences of a multigene cluster-bears striking resemblance to what we have described for the eosinophil-associated ribonuclease genes of the rodent Mus musculus, a finding that may have implications with respect the physiologic function of this unique family of proteins.


Subject(s)
Evolution, Molecular , Rana catesbeiana/genetics , Ribonuclease, Pancreatic/genetics , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Multigene Family , Phylogeny , Rana catesbeiana/physiology , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/classification , Sequence Alignment
13.
Life Sci ; 69(2): 201-12, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11441910

ABSTRACT

Galectin-10 (gal-10, also known as Charcot-Leyden crystal protein) is a member of the galectin family of beta-galactoside binding proteins that is expressed uniquely in eosinophilic and basophilic leukocytes. To gain a better understanding of galectin gene expression, we present an analysis of the transcriptional regulation of the gene encoding gal-10. Analysis of the minimal promoter revealed nine consensus-binding sites for transcription factors, including several that are also found in the minimal promoters of galectins -1, -2, and -3. The decrease in gal-10 promoter activity after disruption of either the GC box (-44 to -50) or the Oct site (-255 to -261) suggests that these sites, along with the previously characterized GATA and EoTF sites, are necessary for full promoter activity. By supershift analysis, we demonstrate binding of the transcription factors Sp1 and Oct1 to the consensus GC box and the Oct site, respectively. Similar to gal-1, gal-10 expression is induced by butyric acid, an effect that is lost upon ablation of the GC box. Additionally, we demonstrate AML3 binding to the consensus AML site and YY1 binding to the Inr sequence, both elements functioning as silencers in the gal-10 promoter.


Subject(s)
Butyric Acid/pharmacology , Eosinophils/metabolism , Gene Expression Regulation , Glycoproteins/genetics , Neoplasm Proteins , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid/genetics , Base Sequence , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Erythroid-Specific DNA-Binding Factors , Glycoproteins/metabolism , HL-60 Cells , Humans , Lysophospholipase , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organic Cation Transporter 1 , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , YY1 Transcription Factor
14.
Gene ; 267(1): 23-30, 2001 Apr 04.
Article in English | MEDLINE | ID: mdl-11311552

ABSTRACT

Mouse eosinophil-associated ribonuclease-2 (mEAR-2) is one of a cluster of genes identified in the genome of the mouse Mus musculus that are highly divergent orthologs of the primate ribonucleases, eosinophil-derived neurotoxin (EDN) and eosinophil cationic protein (ECP). Northern analysis revealed expression of genes hybridizing to mEAR-2 in mouse lung, liver and spleen tissues. We obtained full-length cDNA by hybridization screening of mouse eosinophil and lung cDNA libraries and by rapid amplification of cDNA ends (RACE) from liver, spleen and lung RNA. Using these methods we have isolated the 195 base pair (bp) 3' untranslated region (UTR) that includes a typical polyadenylation signal preceding a poly A tail and the 5' UTR which includes 63-71 bp and three distinct transcriptional start sites. Using unidirectional PCR we isolated a 361-bp 5' promoter region and delineated the intronic / exonic boundaries which include a non-coding exon 1, a single intron, and a coding exon 2, a structure that is typical of genes of the RNase A superfamily. Consensus sites for PU.1 and EoTF, both active as intronic enhancer elements of the gene encoding EDN, are also present in the intron of the gene encoding mEAR-2. The catalytic activity of recombinant baculovirus-derived mEAR-2 is similar to that of rhEDN from this source, with catalytic constants k(cat)/K(m)=5.6x10(6) M(-1) s(-1) and 10.5x10(6) M(-1) s(-1), respectively, against a standard yeast tRNA substrate. Sequence analysis of the non-coding regions and enzymatic characterization of the gene product provide further evidence indicating that mEAR-2 is a structural and functional ortholog of primate EDNs and ECPs.


Subject(s)
Blood Proteins/genetics , Genes/genetics , Animals , Base Sequence , Blood Proteins/metabolism , Blotting, Northern , Cell Line , DNA/chemistry , DNA/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , Eosinophil Granule Proteins , Eosinophil-Derived Neurotoxin , Humans , Mice , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribonucleases/genetics , Ribonucleases/metabolism , Sequence Analysis, DNA , Tissue Distribution , Transcription, Genetic
16.
Cell Immunol ; 213(2): 134-40, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11831875

ABSTRACT

In this work we continue our study of the biochemical responses of respiratory epithelial cells to infection with human paramyxovirus pathogens. In our earlier studies, we detected elevated concentrations of the proinflammatory chemokines MIP-1alpha and IL-8 in upper and lower respiratory tract secretions from patients infected with respiratory syncytial virus (RSV). Here we demonstrate the same trend for individuals infected with parainfluenza virus (PIV), with elevated concentrations of MIP-1alpha and IL-8 (means of 309 +/- 51 and 2280 +/- 440 pg/ml/mg protein, respectively) detected in nasal wash samples from 17 patients with culture-positive PIV. Similar to our findings with RSV, cells of the HEp-2 epithelial line and primary cultures of human bronchial epithelial cells respond to PIV infection with production and release of both MIP-1alpha and IL-8. Addition of the glucocorticoid anti-inflammatory agent hydrocortisone (200-1000 ng/ml) attenuated the production of MIP-1alpha and IL-8 in PIV-infected cells while having minimal to no effect on the production of these mediators from cells infected with RSV. Neither virus infection resulted in a change in the total cellular concentration of glucocorticoid receptors, nor did hydrocortisone exert any differential effect on viral replication. As repression of chemokine production by epithelial cells is likely to result in diminished recruitment of proinflammatory leukocytes, these results may explain in part why glucocorticoid therapy reduces the symptoms associated with acute PIV infection, but have little to no effect in the overall outcome in the case of RSV.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hydrocortisone/pharmacology , Respiratory Syncytial Viruses/physiology , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Child, Preschool , Culture Media , Drug Resistance , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/virology , Female , Gene Expression , Humans , Infant , Interleukin-8/analysis , Interleukin-8/genetics , Macrophage Inflammatory Proteins/analysis , Macrophage Inflammatory Proteins/genetics , Male , Nasal Lavage Fluid/virology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/immunology , Tumor Cells, Cultured , Virus Replication
17.
Genetics ; 156(4): 1949-58, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11102386

ABSTRACT

Host defense against invading pathogens is of great importance to the survival of higher organisms. We have been studying the evolution of mammalian eosinophil-associated ribonucleases (EARs), which are members of the ribonuclease A superfamily with known antipathogen activities. Earlier studies showed that positive selection promoted rapid diversification of paralogous EAR genes in both primates and rodents. Intraspecifically, however, it is unknown whether these genes also have divergent alleles. The recent discovery that the gene repertoire of the EAR family is much larger in rodents than in primates has led us to consider the possibility that primates maintain a large number of polymorphic alleles to compensate for a smaller gene repertoire. Here we present sequences of 2417 nucleotides at the two EAR loci, the eosinophil-derived neurotoxin (EDN, RNase 2) and eosinophil cationic protein (ECP, RNase 3), from >50 human individuals. Our data demonstrate that the nucleotide diversities (0.06-0.11%) at these loci are typical for human nuclear genes, thus permitting us to reject this polymorphism hypothesis. No significant departure from neutrality is noted and no signs of overdominant selection are observed. Similar patterns were observed in a preliminary study of chimpanzees. In summary, our results suggest that the antipathogen functions of the primate EARs are conserved after they are established and that these proteins are not currently undergoing rapid diversification in response to challenge from invading microorganisms.


Subject(s)
Blood Proteins/genetics , Eosinophil Cationic Protein , Evolution, Molecular , Genes , Multigene Family , Proteins/genetics , Ribonucleases , Alleles , Amino Acid Sequence , Animals , Base Sequence , Eosinophil Granule Proteins , Eosinophil-Derived Neurotoxin , Genetic Variation , Haplotypes/genetics , Humans , Infections/enzymology , Infections/genetics , Linkage Disequilibrium , Models, Genetic , Molecular Sequence Data , Pan troglodytes/genetics , Primates/genetics , Rodentia/genetics , Sequence Deletion , Species Specificity , White People/genetics
18.
J Infect Dis ; 182(4): 1022-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10979895

ABSTRACT

We used differential display to detect enhanced expression of an mRNA fragment encoding cytokeratin 17 (Ck-17) in respiratory syncytial virus (RSV)-infected epithelial cells. Expression increased 12-fold by 96 h after infection but remained unchanged in cells challenged with virus in the presence of neutralizing anti-RSV fusion protein antibody. Immunoblots of RSV-infected cell lysates probed with an anti-keratin antibody demonstrated stable expression of total cytokeratins over time. When probed with an anti-Ck-17 monoclonal antibody, Ck-17 was first detected at 4 days after infection. In situ staining demonstrated that Ck-17 expression localized to regions of syncytia formation. Expression of Ck-17 mRNA also increased in response to intracellular RSV-F protein in the absence of active RSV infection. No increase in Ck-17 mRNA expression and no syncytia were observed in RSV-infected cells grown in the presence of the NF-kappaB inhibitor gliotoxin. These results suggest that RSV-induced transcriptional activation of the Ck-17 gene is dependent on an NF-kappaB-associated signaling pathway.


Subject(s)
Giant Cells/physiology , Keratins/genetics , NF-kappa B/metabolism , Respiratory Syncytial Viruses/genetics , Adenoids , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/physiology , Epithelial Cells/virology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Giant Cells/drug effects , Giant Cells/pathology , Gliotoxin/analogs & derivatives , Gliotoxin/pharmacology , Humans , Keratins/biosynthesis , Laryngeal Neoplasms , Recombinant Proteins/biosynthesis , Transfection , Tumor Cells, Cultured
19.
J Immunol ; 165(5): 2677-82, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10946298

ABSTRACT

In this work, we explore the responses of specific gene-deleted mice to infection with the paramyxovirus pneumonia virus of mice (PVM). We have shown previously that infection of wild type mice with PVM results in pulmonary neutrophilia and eosinophilia accompanied by local production of macrophage-inflammatory protein-1 alpha (MIP-1 alpha). Here we examine the role of MIP-1 alpha in the pathogenesis of this disease using mice deficient in MIP-1 alpha or its receptor, CCR1. The inflammatory response to PVM in MIP-1 alpha-deficient mice was minimal, with approximately 10-60 neutrophils/ml and no eosinophils detected in bronchoalveolar lavage fluid. Higher levels of infectious virus were recovered from lung tissue excised from MIP-1 alpha-deficient than from fully competent mice, suggesting that the inflammatory response limits the rate of virus replication in vivo. PVM infection of CCR1-deficient mice was also associated with attenuated inflammation, with enhanced recovery of infectious virus, and with accelerated mortality. These results suggest that the MIP-1 alpha/CCR1-mediated acute inflammatory response protects mice by delaying the lethal sequelae of infection.


Subject(s)
Lung/immunology , Lung/pathology , Macrophage Inflammatory Proteins/physiology , Pneumovirus Infections/immunology , Pneumovirus Infections/pathology , Pneumovirus/immunology , Receptors, Chemokine/physiology , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Movement/immunology , Chemokine CCL4 , Eosinophils/immunology , Eosinophils/pathology , Eosinophils/virology , Female , Lung/metabolism , Lung/virology , Lymphocytes/immunology , Lymphocytes/pathology , Lymphocytes/virology , Macrophage Inflammatory Proteins/deficiency , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/pathology , Neutrophils/virology , Pneumovirus/isolation & purification , Pneumovirus Infections/mortality , Pneumovirus Infections/virology , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/mortality , Pulmonary Eosinophilia/pathology , Receptors, CCR1 , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics
20.
J Leukoc Biol ; 67(5): 691-8, 2000 May.
Article in English | MEDLINE | ID: mdl-10811010

ABSTRACT

The lineage-specific Charcot-Leyden crystal (CLC) protein is found in human eosinophils and basophils where it comprises 7-10% of the cellular protein content. Previous work from our laboratory has identified the motif GGAGA[A/G] as a powerful enhancer of gene transcription ill two eosinophil ribonuclease genes. To evaluate a potentially larger role for this motif in the transcriptional regulation of eosinophil genes, we have isolated 1504 nucleotides 5' to the transcriptional start site of the gene encoding CLC protein and identified a functionally active promoter that includes three distinct copies of the GGAGAA motif. Destruction of only one of the three motifs by site-directed mutagenesis resulted in loss of promoter activity (73 +/- 6% reduction), suggesting that this core motif is necessary but not sufficient to support enhanced transcriptional activity. Sequence comparisons and site-specific mutagenesis has permitted further delineation of this enhancer element which, as a result of this work, is now defined as GGAGA[A/G]NNNA. Electromobility shift assays demonstrated specific binding of nuclear protein(s) from an eosinophilic clone-15 nuclear extract to this extended motif. Similar analysis of a GATA-1 binding site demonstrated enhancer activity, with mutagenesis resulting in a 94 +/- 1.4% reduction in activity, whereas the AML1 site functioned as a gene silencer.


Subject(s)
Basophils/metabolism , Eosinophils/metabolism , Glycoproteins/genetics , Promoter Regions, Genetic , Transcription, Genetic , Base Sequence , Binding Sites , Consensus Sequence , DNA Mutational Analysis , Glycoproteins/blood , Humans , Lysophospholipase , Molecular Sequence Data , Ribonucleases/blood , Ribonucleases/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...