Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Immunity ; 55(3): 494-511.e11, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35263568

ABSTRACT

Interleukin (IL)-22 is central to immune defense at barrier sites. We examined the contributions of innate lymphoid cell (ILC) and T cell-derived IL-22 during Citrobacter rodentium (C.r) infection using mice that both report Il22 expression and allow lineage-specific deletion. ILC-derived IL-22 activated STAT3 in C.r-colonized surface intestinal epithelial cells (IECs) but only temporally restrained bacterial growth. T cell-derived IL-22 induced a more robust and extensive activation of STAT3 in IECs, including IECs lining colonic crypts, and T cell-specific deficiency of IL-22 led to pathogen invasion of the crypts and increased mortality. This reflected a requirement for T cell-derived IL-22 for the expression of a host-protective transcriptomic program that included AMPs, neutrophil-recruiting chemokines, and mucin-related molecules, and it restricted IFNγ-induced proinflammatory genes. Our findings demonstrate spatiotemporal differences in the production and action of IL-22 by ILCs and T cells during infection and reveal an indispensable role for IL-22-producing T cells in the protection of the intestinal crypts.


Subject(s)
Citrobacter rodentium , Enterobacteriaceae Infections , Animals , Anti-Bacterial Agents , Immunity, Innate , Interleukins/metabolism , Intestinal Mucosa , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocytes/metabolism , Interleukin-22
2.
Nat Immunol ; 16(3): 286-95, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25642823

ABSTRACT

Interleukin 17 (IL-17)-producing helper T cells (TH17 cells) and CD4(+) inducible regulatory T cells (iTreg cells) emerge from an overlapping developmental program. In the intestines, the vitamin A metabolite retinoic acid (RA) is produced at steady state and acts as an important cofactor to induce iTreg cell development while potently inhibiting TH17 cell development. Here we found that IL-1 was needed to fully override RA-mediated expression of the transcription factor Foxp3 and induce protective TH17 cell responses. By repressing expression of the negative regulator SOCS3 dependent on the transcription factor NF-κB, IL-1 increased the amplitude and duration of phosphorylation of the transcription factor STAT3 induced by TH17-polarizing cytokines, which led to an altered balance in the binding of STAT3 and STAT5 to shared consensus sequences in developing T cells. Thus, IL-1 signaling modulated STAT activation downstream of cytokine receptors differently to control the TH17 cell-iTreg cell developmental fate.


Subject(s)
Interleukin-1/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/physiology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Tretinoin/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Humans , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation/physiology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism
3.
Eur Respir J ; 60(1)2022 07.
Article in English | MEDLINE | ID: mdl-34916262

ABSTRACT

RATIONALE: The majority of chronic obstructive pulmonary disease (COPD) patients have chronic bronchitis, for which specific therapies are unavailable. Acquired cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction is observed in chronic bronchitis, but has not been proven in a controlled animal model with airway disease. Furthermore, the potential of CFTR as a therapeutic target has not been tested in vivo, given limitations to rodent models of COPD. Ferrets exhibit cystic fibrosis-related lung pathology when CFTR is absent and COPD with bronchitis following cigarette smoke exposure. OBJECTIVES: To evaluate CFTR dysfunction induced by smoking and test its pharmacological reversal by a novel CFTR potentiator, GLPG2196, in a ferret model of COPD with chronic bronchitis. METHODS: Ferrets were exposed for 6 months to cigarette smoke to induce COPD and chronic bronchitis and then treated with enteral GLPG2196 once daily for 1 month. Electrophysiological measurements of ion transport and CFTR function, assessment of mucociliary function by one-micron optical coherence tomography imaging and particle-tracking microrheology, microcomputed tomography imaging, histopathological analysis and quantification of CFTR protein and mRNA expression were used to evaluate mechanistic and pathophysiological changes. MEASUREMENTS AND MAIN RESULTS: Following cigarette smoke exposure, ferrets exhibited CFTR dysfunction, increased mucus viscosity, delayed mucociliary clearance, airway wall thickening and airway epithelial hypertrophy. In COPD ferrets, GLPG2196 treatment reversed CFTR dysfunction, increased mucus transport by decreasing mucus viscosity, and reduced bronchial wall thickening and airway epithelial hypertrophy. CONCLUSIONS: The pharmacologic reversal of acquired CFTR dysfunction is beneficial against pathological features of chronic bronchitis in a COPD ferret model.


Subject(s)
Bronchitis, Chronic , Pulmonary Disease, Chronic Obstructive , Animals , Bronchitis, Chronic/drug therapy , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Ferrets/metabolism , Hypertrophy , Pulmonary Disease, Chronic Obstructive/metabolism , X-Ray Microtomography
4.
Immunity ; 37(6): 1061-75, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23200827

ABSTRACT

Interleukin-22 (IL-22) is central to host protection against bacterial infections at barrier sites. Both innate lymphoid cells (ILCs) and T cells produce IL-22. However, the specific contributions of CD4(+) T cells and their developmental origins are unclear. We found that the enteric pathogen Citrobacter rodentium induced sequential waves of IL-22-producing ILCs and CD4(+) T cells that were each critical to host defense during a primary infection. Whereas IL-22 production by ILCs was strictly IL-23 dependent, development of IL-22-producing CD4(+) T cells occurred via an IL-6-dependent mechanism that was augmented by, but not dependent on, IL-23 and was dependent on both transcription factors T-bet and AhR. Transfer of CD4(+) T cells differentiated with IL-6 in the absence of TGF-ß ("Th22" cells) conferred complete protection of infected IL-22-deficient mice whereas transferred Th17 cells did not. These findings establish Th22 cells as an important component of mucosal antimicrobial host defense.


Subject(s)
Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Interleukins/physiology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Enterobacteriaceae Infections/mortality , Enterobacteriaceae Infections/prevention & control , Gene Expression Regulation , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interleukin-23/immunology , Interleukin-23/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Interleukins/metabolism , Mice , Mice, Knockout , Mucous Membrane/immunology , Mucous Membrane/microbiology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Interleukin-22
5.
J Immunol ; 202(4): 1039-1044, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30642977

ABSTRACT

The ICOS pathway has been implicated in the development and functions of regulatory T (Treg) cells, including those producing IL-10. Treg cell-derived IL-10 is indispensable for the establishment and maintenance of intestinal immune homeostasis. We examined the possible involvement of the ICOS pathway in the accumulation of murine colonic Foxp3- and/or IL-10-expressing cells. We show that ICOS deficiency does not impair induction of IL-10 by intestinal CD4 T cells but, instead, triggers substantial reductions in gut-resident and peripherally derived Foxp3+ Treg cells. ICOS deficiency is associated with reduced demethylation of Foxp3 CNS2 and enhanced loss of Foxp3. This instability significantly limits the ability of ICOS-deficient Treg cells to reverse ongoing inflammation. Collectively, our results identify a novel role for ICOS costimulation in imprinting the functional stability of Foxp3 that is required for the retention of full Treg cell function in the periphery.


Subject(s)
Down-Regulation , Forkhead Transcription Factors/metabolism , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukin-10/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Down-Regulation/immunology , Forkhead Transcription Factors/immunology , Inducible T-Cell Co-Stimulator Protein/deficiency , Inducible T-Cell Co-Stimulator Protein/immunology , Inflammation/immunology , Inflammation/metabolism , Interleukin-10/immunology , Mice , Mice, Knockout , Mice, Transgenic , T-Lymphocytes, Regulatory/immunology
6.
Infect Immun ; 88(4)2020 03 23.
Article in English | MEDLINE | ID: mdl-31932329

ABSTRACT

Stenotrophomonas maltophilia is a Gram-negative bacterium found ubiquitously in the environment that has historically been regarded as nonpathogenic. S. maltophilia is increasingly observed in patient sputa in cystic fibrosis (CF), and while existing epidemiology indicates that patients with S. maltophilia have poorer diagnoses, its clinical significance remains unclear. Moreover, as multidrug resistance is common among S. maltophilia isolates, treatment options for these infections may be limited. Here, we investigated the pathogenicity of S. maltophilia alone and during polymicrobial infection with Pseudomonas aeruginosa Colonization, persistence, and virulence of S. maltophilia were assessed in experimental respiratory infections of mice. The results of this study indicate that S. maltophilia transiently colonizes the lung accompanied by significant weight loss and immune cell infiltration and the expression of early inflammatory markers, including interleukin 6 (IL-6), IL-1α, and tumor necrosis factor alpha (TNF-α). Importantly, polymicrobial infection with P. aeruginosa elicited significantly higher S. maltophilia counts in bronchoalveolar lavages and lung tissue homogenates. This increase in bacterial load was directly correlated with the density of the P. aeruginosa population and required viable P. aeruginosa bacteria. Microscopic analysis of biofilms formed in vitro revealed that S. maltophilia formed well-integrated biofilms with P. aeruginosa, and these organisms colocalize in the lung during dual-species infection. Based on these results, we conclude that active cellular processes by P. aeruginosa afford a significant benefit to S. maltophilia during polymicrobial infections. Furthermore, these results indicate that S. maltophilia may have clinical significance in respiratory infections.


Subject(s)
Coinfection/microbiology , Gram-Negative Bacterial Infections/microbiology , Microbial Interactions , Pneumonia, Bacterial/microbiology , Pseudomonas aeruginosa/growth & development , Stenotrophomonas maltophilia/growth & development , Animals , Bacterial Load , Body Weight , Bronchoalveolar Lavage Fluid/microbiology , Coinfection/pathology , Disease Models, Animal , Gram-Negative Bacterial Infections/pathology , Immunity, Innate , Lung/microbiology , Lung/pathology , Mice , Pneumonia, Bacterial/pathology
7.
Am J Physiol Lung Cell Mol Physiol ; 319(1): L11-L20, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32374671

ABSTRACT

Structural changes to airway morphology, such as increased bronchial wall thickness (BWT) and airway wall area, are cardinal features of chronic obstructive pulmonary disease (COPD). Ferrets are a recently established animal model uniquely exhibiting similar clinical and pathological characteristics of COPD as humans, including chronic bronchitis. Our objective was to develop a microcomputed tomography (µCT) method for evaluating structural changes to the airways in ferrets and assess whether the effects of smoking induce changes consistent with chronic bronchitis in humans. Ferrets were exposed to mainstream cigarette smoke or air control twice daily for 6 mo. µCT was conducted in vivo at 6 mo; a longitudinal cohort was imaged monthly. Manual measurements of BWT, luminal diameter (LD), and BWT-to-LD ratio (BWT/LD) were conducted and confirmed by a semiautomated algorithm. The square root of bronchial wall area (√WA) versus luminal perimeter was determined on an individual ferret basis. Smoke-exposed ferrets reproducibly demonstrated 34% increased BWT (P < 0.001) along with increased LD and BWT/LD versus air controls. Regression indicated that the effect of smoking on BWT persisted despite controlling for covariates. Semiautomated measurements replicated findings. √WA for the theoretical median airway luminal perimeter of 4 mm (Pi4) was elevated 4.4% in smoke-exposed ferrets (P = 0.015). Increased BWT and Pi4 developed steadily over time. µCT-based airway measurements in ferrets are feasible and reproducible. Smoke-exposed ferrets develop increased BWT and Pi4, changes similar to humans with chronic bronchitis. µCT can be used as a significant translational platform to measure dynamic airway morphological changes.

8.
J Allergy Clin Immunol ; 143(3): 1183-1197.e7, 2019 03.
Article in English | MEDLINE | ID: mdl-30092287

ABSTRACT

BACKGROUND: Mycoplasma pneumoniae, an atypical human pathogen, has been associated with asthma initiation and exacerbation. Asthmatic patients have been reported to have higher carriage rates of M pneumoniae compared with nonasthmatic subjects and are at greater risk for invasive respiratory infections. OBJECTIVE: We sought to study whether prior allergen sensitization affects the host response to chronic bacterial infection. METHODS: BALB/cJ and IL-4 receptor α-/- mice were sensitized with ovalbumin (OVA) and then infected with M pneumoniae or Streptococcus pneumoniae. Immune parameters were analyzed at 30 days postinfection and included cellular profiles in bronchoalveolar lavage fluid (BALF) and serum IgG and IgE antibody levels to whole bacterial lysate, recombinant P1 adhesin, and OVA. Total lung RNA was examined for transcript levels, and BALF was examined for cytokine protein profiles. RESULTS: Anti-M pneumoniae antibody responses were decreased in allergen-sensitized, M pneumoniae-infected animals compared with control animals, but OVA-specific IgG responses were unaffected. Similar decreases in anti-S pneumoniae antibody levels were found in OVA-sensitized animals. However, M pneumoniae, but not S pneumoniae, infection augmented anti-OVA IgE antibody responses. Loss of IL-4 receptor signaling partially restored anti-M pneumoniae antibody responses in IgG2a and IgG2b subclasses. Inflammatory cytokine levels in BALF from OVA-sensitized, M pneumoniae-infected or S pneumoniae-infected animals were reduced compared with those in uninfected OVA-sensitized control animals. Unexpectedly, airway hyperreactivity to methacholine was essentially ablated in M pneumoniae-infected, OVA-sensitized animals. CONCLUSIONS: An established type 2-biased host immune response impairs the host immune response to respiratory bacterial infection in a largely pathogen-independent manner. Some pathogens, such as M pneumoniae, can augment ongoing allergic responses and inhibit pulmonary type 2 cytokine responses and allergic airway hyperreactivity.


Subject(s)
Asthma/immunology , Immunoglobulin G/immunology , Pneumococcal Infections/immunology , Pneumonia, Mycoplasma/immunology , Respiratory Tract Infections/immunology , Allergens/immunology , Animals , Asthma/pathology , Asthma/physiopathology , Cytokines/genetics , Lung/pathology , Lung/physiopathology , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin/immunology , Pneumococcal Infections/pathology , Pneumococcal Infections/physiopathology , Pneumonia, Mycoplasma/pathology , Pneumonia, Mycoplasma/physiopathology , Receptors, Cell Surface/genetics , Respiratory Tract Infections/pathology , Respiratory Tract Infections/physiopathology
9.
Genes Immun ; 20(2): 158-166, 2019 02.
Article in English | MEDLINE | ID: mdl-29599513

ABSTRACT

Studies have identified abnormalities in the microbiota of patients with arthritis. To evaluate the pathogenicity of human microbiota, we performed fecal microbial transplantation from children with spondyloarthritis and controls to germ-free KRN/B6xNOD mice. Ankle swelling was equivalent in those that received patient vs. control microbiota. Principal coordinates analysis revealed incomplete uptake of the human microbiota with over-representation of two genera (Bacteroides and Akkermansia) among the transplanted mice. The microbiota predicted the extent of ankle swelling (R2 = 0.185, p = 0.018). The abundances of Bacteroides (r = -0.510, p = 0.010) inversely and Akkermansia (r = 0.367, p = 0.078) directly correlated with ankle swelling. Addition of Akkermansia muciniphila to Altered Schaedler's Flora (ASF) resulted in small but statistically significant increased ankle swelling as compared to mice that received ASF alone (4.0 mm, 3.9-4.1 vs. 3.9 mm, IQR 3.6-4.0, p = 0.041), as did addition of A. muciniphila cultures to transplanted human microbiota as compared to mice that received transplanted human microbiota alone (4.5 mm, IQR 4.3-5.5 vs. 4.1 mm, IQR 3.9-4.3, p = 0.019). This study supports previous findings of an association between A. muciniphila and arthritis.


Subject(s)
Arthritis/microbiology , Gastrointestinal Microbiome , Adolescent , Animals , Ankle/pathology , Bacteroides/isolation & purification , Bacteroides/pathogenicity , Child , Female , Humans , Male , Mice , Mice, Inbred NOD , Verrucomicrobia/isolation & purification , Verrucomicrobia/pathogenicity
10.
Am J Physiol Renal Physiol ; 316(3): F463-F472, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30600684

ABSTRACT

Heterozygosity for human polycystic kidney and hepatic disease 1 ( PKHD1) mutations was recently associated with cystic liver disease and radiographic findings resembling medullary sponge kidney (MSK). However, the relevance of these associations has been tempered by a lack of cystic liver or renal disease in heterozygous mice carrying Pkhd1 gene trap or exon deletions. To determine whether heterozygosity for a smaller Pkhd1 defect can trigger cystic renal disease in mice, we generated and characterized mice with the predicted truncating Pkhd1C642* mutation in a region corresponding to the middle of exon 20 cluster of five truncating human mutations (between PKHD1G617fs and PKHD1G644*). Mouse heterozygotes or homozygotes for the Pkhd1C642* mutation did not have noticeable liver or renal abnormalities on magnetic resonance images during their first weeks of life. However, when aged to ~1.5 yr, the Pkhd1C642* heterozygotes developed prominent cystic liver changes; tissue analyses revealed biliary cysts and increased number of bile ducts without signs of congenital hepatic fibrosis-like portal field inflammation and fibrosis that was seen in Pkhd1C642* homozygotes. Interestingly, aged female Pkhd1C642* heterozygotes, as well as homozygotes, developed radiographic changes resembling MSK. However, these changes correspond to proximal tubule ectasia, not an MSK-associated collecting duct ectasia. In summary, by demonstrating that cystic liver and kidney abnormalities are triggered by heterozygosity for the Pkhd1C642* mutation, we provide important validation for relevant human association studies. Together, these investigations indicate that PKHD1 mutation heterozygosity (predicted frequency 1 in 70 individuals) is an important underlying cause of cystic liver disorders and MSK-like manifestations in a human population.


Subject(s)
Cysts/diagnostic imaging , Kidney Diseases/diagnostic imaging , Kidney Tubules, Proximal/diagnostic imaging , Liver Diseases/diagnostic imaging , Medullary Sponge Kidney/diagnostic imaging , Receptors, Cell Surface/metabolism , Animals , Cysts/genetics , Cysts/metabolism , Diagnosis, Differential , Dilatation, Pathologic/diagnostic imaging , Dilatation, Pathologic/genetics , Dilatation, Pathologic/metabolism , Disease Models, Animal , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Tubules, Proximal/metabolism , Liver Diseases/genetics , Liver Diseases/metabolism , Magnetic Resonance Imaging , Medullary Sponge Kidney/genetics , Medullary Sponge Kidney/metabolism , Mice , Mice, Knockout , Receptors, Cell Surface/genetics
11.
Mamm Genome ; 30(9-10): 245-259, 2019 10.
Article in English | MEDLINE | ID: mdl-31673770

ABSTRACT

Genome-wide association studies (GWAS) and functional genomic analyses have implicated several ITGAM (CD11b) single-nucleotide polymorphisms (SNPs) in the development of SLE and other disorders. ITGAM encodes the αM chain of the ß2 integrin Mac-1, a receptor that plays important roles in myeloid cell functions. The ITGAM SNP rs1143679, which results in an arginine to histidine change at amino acid position 77 of the CD11b protein, has been shown to reduce binding to several ligands and to alter Mac-1-mediated cellular response in vitro. Importantly, however, the potential contribution of this SNP variant to the initiation and/or progression of immune and inflammatory processes in vivo remains unexplored. Herein, we describe for the first time the generation and characterization of a mouse line expressing the 77His variant of CD11b. Surprisingly, we found that 77His did not significantly affect Mac-1-mediated leukocyte migration and activation as assessed using thioglycollate-induced peritonitis and LPS/TNF-α-induced dermal inflammation models. In contrast, expression of this variant did alter T cell immunity, as evidenced by significantly reduced proliferation of ovalbumin (OVA)-specific transgenic T cells in 77His mice immunized with OVA. Reduced antigen-specific T cell proliferation was also observed when either 77His splenic dendritic cells (DCs) or bone marrow-derived DCs were used as antigen-presenting cells (APCs). Although more work is necessary to determine how this alteration might influence the development of SLE or other diseases, these in vivo findings suggest that the 77His variant of CD11b can compromise the ability of DCs to induce antigen-driven T cell proliferation.


Subject(s)
CD11b Antigen/genetics , Dendritic Cells/immunology , Polymorphism, Single Nucleotide , T-Lymphocytes/cytology , Alleles , Amino Acid Substitution , Animals , CD11b Antigen/immunology , Cell Proliferation , Female , Genome-Wide Association Study , Genotype , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology
12.
J Immunol ; 199(1): 312-322, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28539431

ABSTRACT

Th17 cells play a role as an inflammation mediator in a variety of autoimmune disorders, including inflammatory bowel disease, and thus are widely considered to be pathogenic. However, Th17 cells are present in the normal intestine and show a homeostatic phenotype; that is, they participate in the maintenance of intestinal homeostasis rather than inducing inflammation. We observed an enlarged Th17 population in the small intestine of C57BL/6.IgA-/- mice compared with wild-type mice, which was further amplified with cholera toxin (CT) immunization without causing intestinal inflammation. The increased Th17 induction and the correspondingly 10-fold higher CT B subunit-specific serum IgG response in IgA-/- mice after CT immunization was microbiota dependent and was associated with increased segmented filamentous bacteria in the small intestine of IgA-/- mice. Oral administration of vancomycin greatly dampened both CT immunogenicity and adjuvanticity, and the differential CT responses in IgA-/- and wild-type mice disappeared after intestinal microbiota equalization. Using gnotobiotic mouse models, we found that CT induction of homeostatic intestinal Th17 responses was supported not only by segmented filamentous bacteria, but also by other commensal bacteria. Furthermore, transcriptome analysis using IL-17AhCD2 reporter mice revealed a similar gene expression profile in CT-induced intestinal Th17 cells and endogenous intestinal Th17 cells at homeostasis, with upregulated expression of a panel of immune-regulatory genes, which was distinctly different from the gene expression profile of pathogenic Th17 cells. Taken together, we identified a nonpathogenic signature of intestinal homeostatic Th17 cells, which are actively regulated by the commensal microbiota and can be selectively stimulated by CT.


Subject(s)
Cholera Toxin/immunology , Gastrointestinal Microbiome/immunology , Homeostasis , Intestinal Mucosa/immunology , Intestine, Small/immunology , Th17 Cells/immunology , Animals , Cholera Toxin/administration & dosage , Gastrointestinal Microbiome/physiology , Gene Expression Profiling , Germ-Free Life , IgA Deficiency/immunology , Immunoglobulin A/immunology , Inflammatory Bowel Diseases , Intestinal Mucosa/microbiology , Intestine, Small/microbiology , Mice , Mice, Inbred C57BL , Vancomycin/administration & dosage
13.
Am J Physiol Renal Physiol ; 314(6): F1166-F1176, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29361668

ABSTRACT

Lewisite (2-chlorovinyldichloroarsine) is an organic arsenical chemical warfare agent that was developed and weaponized during World Wars I/II. Stockpiles of lewisite still exist in many parts of the world and pose potential environmental and human health threat. Exposure to lewisite and similar chemicals causes intense cutaneous inflammatory response. However, morbidity and mortality in the exposed population is not only the result of cutaneous damage but is also a result of systemic injury. Here, we provide data delineating the pathogenesis of acute kidney injury (AKI) following cutaneous exposure to lewisite and its analog phenylarsine oxide (PAO) in a murine model. Both agents caused renal tubular injury, characterized by loss of brush border in proximal tubules and tubular cell apoptosis accompanied by increases in serum creatinine, neutrophil gelatinase-associated lipocalin, and kidney injury molecule-1. Interestingly, lewisite exposure enhanced production of reactive oxygen species (ROS) in the kidney and resulted in the activation of autophagic and DNA damage response (DDR) signaling pathways with increased expression of beclin-1, autophagy-related gene 7, and LC-3A/B-II and increased phosphorylation of γ-H2A.X and checkpoint kinase 1/2, respectively. Terminal deoxyribonucleotide-transferase-mediated dUTP nick-end labeling-positive cells were detected in renal tubules along with enhanced proapoptotic BAX/cleaved caspase-3 and reduced antiapoptotic BCL2. Scavenging ROS by cutaneous postexposure application of the antioxidant N-acetyl-l-cysteine reduced lewisite-induced autophagy and DNA damage. In summary, we provide evidence that topical exposure to lewisite causes AKI. The molecular mechanism underlying these changes involves ROS-dependent activation of autophagy and DDR pathway associated with the induction of apoptosis.


Subject(s)
Acute Kidney Injury/chemically induced , Arsenicals/adverse effects , Autophagy , Chemical Warfare Agents/adverse effects , DNA Damage , Kidney/pathology , Skin Absorption , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Arsenicals/metabolism , Chemical Warfare Agents/metabolism , Cytokines/metabolism , Female , HEK293 Cells , Humans , Kidney/metabolism , Male , Mice, Hairless , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
14.
Am J Transplant ; 18(11): 2647-2662, 2018 11.
Article in English | MEDLINE | ID: mdl-29659179

ABSTRACT

Human cytomegalovirus (HCMV) donor positive (D+) serostatus with acute rejection is associated with renal allograft loss, but the impact of recipient positive (R+) serostatus is unclear. In an allogeneic renal transplant model, antiviral natural killer (NK) and CD8+ T cell memory responses in murine CMV (MCMV) D+/R+ transplants were compared to D-/R- and D+/R- transplants, with recipient infection varied by MCMV dose and strain. D+/R- transplants had high primary antiviral cytolytic (interferon-γ+) and cytotoxic (granzyme B+) NK responses, whereas NK memory responses were lower in D+/R+ recipients receiving a high primary MCMV dose. Despite MCMV immunity, D+/R+ recipients receiving a low MCMV dose showed primary-like high cytolytic and cytotoxic NK responses. D+/R+ transplants infected with different D/R strains had low cytolytic NK responses but high cytotoxic NK responses. NK memory also induced a novel TNF-α+ NK response among high-dose virus recipients. MCMV+ transplants had greater Th17 responses than MCMV-uninfected transplants and Th17 inhibition ameliorated graft injury. All MCMV+ recipients had similar CD8+ T cell responses. In sum, NK and Th17 responses, but not CD8+ T cells, varied according to conditions of primary recipient infection. This variability could contribute to variable graft outcomes in HCMV D+/R+ renal transplantation.


Subject(s)
Cytomegalovirus Infections/immunology , Graft Rejection/etiology , Kidney Transplantation/adverse effects , Killer Cells, Natural/immunology , Muromegalovirus/classification , Th17 Cells/immunology , Viral Load/immunology , Allografts , Animals , Cytomegalovirus Infections/virology , Graft Rejection/pathology , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muromegalovirus/immunology , Th17 Cells/pathology , Th17 Cells/virology
15.
Proc Natl Acad Sci U S A ; 112(22): 7061-6, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-26038559

ABSTRACT

Th17 cells reactive to the enteric microbiota are central to the pathogenesis of certain types of inflammatory bowel disease. However, Th17 cells display substantial developmental plasticity, such that some progeny of Th17 cell precursors retain a predominantly IL-17A(+) phenotype, whereas others extinguish IL-17 expression and acquire expression of IFN-γ, giving rise to "Th1-like" cells. It remains unclear what role these subsets play in inflammatory bowel disease. Using a Th17 transfer model of colitis, we found that IFN-γ-deficient Th17 cells retained an IL-17A(+) phenotype and were unable to induce colitis in recipients. Development of disease required the transition of a subset of Th17 precursors to Th1-like cells and was contingent on the expression of both Stat4 and T-bet, but not the IL-12 or IFN-γ receptors. Moreover, Th17 cells could provide "help" for the development of pathogenic Th1 cells from naïve precursors. These results indicate that Th17 cells are potent mediators of colitis pathogenesis by dual mechanisms: by directly transitioning to Th1-like cells and by supporting the development of classic Th1 cells.


Subject(s)
Cell Differentiation/immunology , Inflammatory Bowel Diseases/physiopathology , Th1 Cells/immunology , Th17 Cells/cytology , Adoptive Transfer , Animals , Inflammatory Bowel Diseases/immunology , Interferon-gamma/immunology , Mice , Mice, Knockout , STAT4 Transcription Factor/metabolism , Statistics, Nonparametric , T-Box Domain Proteins/metabolism , Th17 Cells/immunology
16.
J Immunol ; 195(10): 4685-98, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26466956

ABSTRACT

Apolipoprotein (Apo)A-I, the major lipid-binding protein of high-density lipoprotein, can prevent autoimmunity and suppress inflammation in hypercholesterolemic mice by attenuating lymphocyte cholesterol accumulation and removing tissue-oxidized lipids. However, whether ApoA-I mediates immune-suppressive or anti-inflammatory effects under normocholesterolemic conditions and the mechanisms involved remain unresolved. We transferred bone marrow from systemic lupus erythematosus (SLE)-prone Sle123 mice into normal, ApoA-I-knockout (ApoA-I(-/-)) and ApoA-I-transgenic (ApoA-I(tg)) mice. Increased ApoA-I in ApoA-I(tg) mice suppressed CD4(+) T and B cell activation without changing lymphocyte cholesterol levels or reducing major ApoA-I-binding oxidized fatty acids. Unexpectedly, oxidized fatty acid peroxisome proliferator-activated receptor γ ligands 13- and 9-hydroxyoctadecadienoic acid were increased in lymphocytes of autoimmune ApoA-I(tg) mice. ApoA-I reduced Th1 cells independently of changes in CD4(+)Foxp3(+) regulatory T cells or CD11c(+) dendritic cell activation and migration. Follicular helper T cells, germinal center B cells, and autoantibodies were also lower in ApoA-I(tg) mice. Transgenic ApoA-I also improved SLE-mediated glomerulonephritis. However, ApoA-I deficiency did not have the opposite effects on autoimmunity or glomerulonephritis, possibly as the result of compensatory increases in ApoE on high-density lipoprotein. We conclude that, although compensatory mechanisms prevent the proinflammatory effects of ApoA-I deficiency in normocholesterolemic mice, increasing ApoA-I can attenuate lymphocyte activation and autoimmunity in SLE independently of cholesterol transport, possibly through oxidized fatty acid peroxisome proliferator-activated receptor γ ligands, and it can reduce renal inflammation in glomerulonephritis.


Subject(s)
Apolipoprotein A-I/metabolism , Autoimmunity/immunology , Cholesterol/metabolism , Lupus Nephritis/immunology , Lymphocyte Activation/immunology , Animals , Apolipoprotein A-I/genetics , Apolipoprotein A-I/immunology , Apolipoproteins E/metabolism , Autoantibodies/blood , Autoantibodies/immunology , Autoimmunity/genetics , B-Lymphocytes/immunology , Bone Marrow Transplantation , Cell Movement/immunology , Dendritic Cells/immunology , Gas Chromatography-Mass Spectrometry , Linoleic Acids/metabolism , Lipoproteins, HDL/immunology , Lupus Nephritis/genetics , Lupus Nephritis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/metabolism , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology
17.
Appl Environ Microbiol ; 82(23): 6952-6960, 2016 12.
Article in English | MEDLINE | ID: mdl-27663026

ABSTRACT

Colonization with Oxalobacter formigenes may reduce the risk of calcium oxalate kidney stone disease. To improve our limited understanding of host/O.formigenes and microbe/O.formigenes interactions, germ-free or altered Schaedler flora (ASF) mice were colonized with O.formigenes Germ-free mice were stably colonized with O.formigenes suggesting O.formigenes does not require other organisms to sustain its survival. Examination of intestinal material indicated no viable O.formigenes in the small intestine, ∼4 × 106 O.formigenes per 100mg contents in the cecum and proximal colon, and ∼0.02% of total cecal O. formigenes cells were tightly associated to the mucosa. O.formigenes did not alter the overall microbial composition of ASF, and ASF did not impact O.formigenes capacity to degrade dietary oxalate in the cecum. 24-hour urine and fecal collections within metabolic cages in semi-rigid isolators demonstrated that introduction of ASF into germ-free mice significantly reduced urinary oxalate excretion. These experiments also showed that mono-colonized O.formigenes mice excrete significantly more urinary calcium compared to germ-free mice, which may be due to degradation of calcium oxalate crystals by O.formigenes and the subsequent intestinal absorption of free calcium. In conclusion, the successful establishment of defined-flora O.formigenes mouse models should improve our understanding of O.formigenes host and microbe interactions. These data support the use of O.formigenes as a probiotic that has limited impact on the composition of the resident microbiota but providing efficient oxalate degrading function. IMPORTANCE: Despite evidence suggesting a lack of O. formigenes colonization is a risk factor for calcium oxalate stone formation, little is known about O. formigenes biology. This study is the first to utilize germ-free mice to examine the response to mono-colonization with O. formigenes and the impact of a defined bacterial cocktail, altered Schaedler flora, on O. formigenes colonization. This study demonstrates that germ-free mice on their regular diet remain mono-colonized with O. formigenes, and suggests that further studies with O. formigenes gnotobiotic mouse models could improve our understanding of O. formigenes biology and host/O. formigenes and microbe/O. formigenes interactions.

18.
J Immunol ; 193(3): 1353-63, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24973442

ABSTRACT

Mycoplasmas are a common cause of pneumonia in humans and animals, and attempts to create vaccines have not only failed to generate protective host responses, but they have exacerbated the disease. Mycoplasma pulmonis causes a chronic inflammatory lung disease resulting from a persistent infection, similar to other mycoplasma respiratory diseases. Using this model, Th1 subsets promote resistance to mycoplasma disease and infection, whereas Th2 responses contribute to immunopathology. The purpose of the present study was to evaluate the capacity of cytokine-differentiated dendritic cell (DC) populations to influence the generation of protective and/or pathologic immune responses during M. pulmonis respiratory disease in BALB/c mice. We hypothesized that intratracheal inoculation of mycoplasma Ag-pulsed bone marrow-derived DCs could result in the generation of protective T cell responses during mycoplasma infection. However, intratracheal inoculation (priming) of mice with Ag-pulsed DCs resulted in enhanced pathology in the recipient mice when challenged with mycoplasma. Inoculation of immunodeficient SCID mice with Ag-pulsed DCs demonstrated that this effect was dependent on lymphocyte responses. Similar results were observed when mice were primed with Ag-pulsed pulmonary, but not splenic, DCs. Lymphocytes generated in uninfected mice after the transfer of either Ag-pulsed bone marrow-derived DCs or pulmonary DCs were shown to be IL-13(+) Th2 cells, known to be associated with immunopathology. Thus, resident pulmonary DCs most likely promote the development of immunopathology in mycoplasma disease through the generation of mycoplasma-specific Th2 responses. Vaccination strategies that disrupt or bypass this process could potentially result in a more effective vaccination.


Subject(s)
Antigens, Bacterial/administration & dosage , Bone Marrow Cells/immunology , Dendritic Cells/immunology , Lung/immunology , Mycoplasma pulmonis/immunology , Pneumonia, Mycoplasma/immunology , Th2 Cells/immunology , Administration, Intranasal , Animals , Bone Marrow Cells/microbiology , Bone Marrow Cells/pathology , Dendritic Cells/pathology , Dendritic Cells/transplantation , Female , Intubation, Intratracheal , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, SCID , Mycoplasma pulmonis/pathogenicity , Pneumonia, Mycoplasma/microbiology , Pneumonia, Mycoplasma/pathology , Th2 Cells/pathology , Th2 Cells/transplantation
19.
Development ; 138(9): 1675-85, 2011 May.
Article in English | MEDLINE | ID: mdl-21429982

ABSTRACT

Skin and hair follicle morphogenesis and homeostasis require the integration of multiple signaling pathways, including Hedgehog (Hh) and Wingless (Wnt), and oriented cell divisions, all of which have been associated with primary cilia. Although studies have shown that disrupting dermal cilia causes follicular arrest and attenuated Hh signaling, little is known about the role of epidermal cilia. Here, epidermal cilia function was analyzed using conditional alleles of the ciliogenic genes Ift88 and Kif3a. At birth, epidermal cilia mutants appeared normal, but developed basaloid hyperplasia and ingrowths into the dermis of the ventrum with age. In addition, follicles in the tail were disorganized and had excess sebaceous gland lobules. Epidermal cilia mutants displayed fewer long-term label-retaining cells, suggesting altered stem cell homeostasis. Abnormal proliferation and differentiation were evident from lineage-tracing studies and showed an expansion of follicular cells into the interfollicular epidermis, as is seen during wound repair. These phenotypes were not associated with changes in canonical Wnt activity or oriented cell division. However, nuclear accumulation of the ΔNp63 transcription factor, which is involved in stratification, keratinocyte differentiation and wound repair, was increased, whereas the Hh pathway was repressed. Intriguingly, the phenotypes were not typical of those associated with loss of Hh signaling but exhibited similarities with those of mice in which ΔNp63 is overexpressed in the epidermis. Collectively, these data indicate that epidermal primary cilia may function in stress responses and epidermal homeostasis involving pathways other than those typically associated with primary cilia.


Subject(s)
Cilia/physiology , Epidermal Cells , Hair Follicle/physiology , Homeostasis/physiology , Skin Physiological Phenomena , Animals , Animals, Newborn , Cilia/genetics , Cilia/metabolism , Epidermis/metabolism , Epidermis/physiology , Gene Expression Regulation, Developmental , Hair Follicle/cytology , Hair Follicle/metabolism , Homeostasis/genetics , Integrases/genetics , Integrases/metabolism , Kinesins/genetics , Kinesins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Skin Physiological Phenomena/genetics , Transgenes/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
20.
Mol Genet Metab ; 111(3): 374-381, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24411223

ABSTRACT

Nonsense suppression therapy is a therapeutic approach aimed at treating genetic diseases caused by in-frame premature termination codons (PTCs; also commonly known as nonsense mutations). This approach utilizes compounds that suppress translation termination at PTCs, which allows translation to continue and partial levels of deficient protein function to be restored. We hypothesize that suppression therapy can attenuate the lysosomal storage disease mucopolysaccharidosis type I-Hurler (MPS I-H), the severe form of α-L-iduronidase deficiency. α-L-iduronidase participates in glycosaminoglycan (GAG) catabolism and its insufficiency causes progressive GAG accumulation and onset of the MPS I-H phenotype, which consists of multiple somatic and neurological defects. 60-80% of MPS I-H patients carry a nonsense mutation in the IDUA gene. We previously showed that 2-week treatment with the designer aminoglycoside NB84 restored enough α-L-iduronidase function via PTC suppression to reduce tissue GAG accumulation in the Idua(tm1Kmke) MPS I-H mouse model, which carries a PTC homologous to the human IDUA-W402X nonsense mutation. Here we report that long-term NB84 administration maintains α-L-iduronidase activity and GAG reduction in Idua(tm1Kmke) mice throughout a 28-week treatment period. An examination of more complex MPS I-H phenotypes in Idua(tm1Kmke) mice following 28-week NB84 treatment revealed significant moderation of the disease in multiple tissues, including the brain, heart and bone, that are resistant to current MPS I-H therapies. This study represents the first demonstration that long-term nonsense suppression therapy can moderate progression of a genetic disease.


Subject(s)
Aminoglycosides/administration & dosage , Codon, Nonsense/genetics , Iduronidase/genetics , Mucopolysaccharidosis I/genetics , Trisaccharides/administration & dosage , Animals , Disease Models, Animal , Disease Progression , Glycosaminoglycans/metabolism , Humans , Iduronidase/metabolism , Mice , Mucopolysaccharidosis I/drug therapy , Mucopolysaccharidosis I/enzymology , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL