Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(51): e2211534119, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36508653

ABSTRACT

Food fortification is an effective strategy to address vitamin A (VitA) deficiency, which is the leading cause of childhood blindness and drastically increases mortality from severe infections. However, VitA food fortification remains challenging due to significant degradation during storage and cooking. We utilized an FDA-approved, thermostable, and pH-responsive basic methacrylate copolymer (BMC) to encapsulate and stabilize VitA in microparticles (MPs). Encapsulation of VitA in VitA-BMC MPs greatly improved stability during simulated cooking conditions and long-term storage. VitA absorption was nine times greater from cooked MPs than from cooked free VitA in rats. In a randomized controlled cross-over study in healthy premenopausal women, VitA was readily released from MPs after consumption and had a similar absorption profile to free VitA. This VitA encapsulation technology will enable global food fortification strategies toward eliminating VitA deficiency.


Subject(s)
Vitamin A Deficiency , Vitamin A , Female , Rats , Animals , Food, Fortified , Cross-Over Studies , Cooking , Micronutrients
2.
Bioconjug Chem ; 34(1): 193-203, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36580277

ABSTRACT

Recently, there has been increased interest in using mannan as an immunomodulatory bioconjugate. Despite notable immunological and functional differences between the reduced (R-Man) and oxidized (O-Man) forms of mannan, little is known about the impact of mannan oxidation state on its in vivo persistence or its potential controlled release from biomaterials that may improve immunotherapeutic or prophylactic efficacy. Here, we investigate the impact of oxidation state on the in vitro and in vivo release of mannan from a biocompatible and immunostimulatory multidomain peptide hydrogel, K2(SL)6K2 (abbreviated as K2), that has been previously used for the controlled release of protein and small molecule payloads. We observed that O-Man released more slowly from K2 hydrogels in vitro than R-Man. In vivo, the clearance of O-Man from K2 hydrogels was slower than O-Man alone. We attributed the slower release rate to the formation of dynamic imine bonds between reactive aldehyde groups on O-Man and the lysine residues on K2. This imine interaction was also observed to improve K2 + O-Man hydrogel strength and shear recovery without significantly influencing secondary structure or peptide nanofiber formation. There were no observed differences in the in vivo release rates of O-Man loaded in K2, R-Man loaded in K2, and R-Man alone. These data suggest that, after subcutaneous injection, R-Man naturally persists longer in vivo than O-Man and minimally interacts with the peptide hydrogel. These results highlight a potentially critical, but previously unreported, difference in the in vivo behavior of O-Man and R-Man and demonstrate that K2 can be used to normalize the release of O-Man to that of R-Man. Further, since K2 itself is an adjuvant, a combination of O-Man and K2 could be used to enhance the immunostimulatory effects of O-Man for applications such as infectious disease vaccines and cancer immunotherapy.


Subject(s)
Nanofibers , Humans , Nanofibers/chemistry , Mannans , Delayed-Action Preparations , Hydrogels/chemistry , Peptides/chemistry
3.
Proc Natl Acad Sci U S A ; 115(23): E5269-E5278, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29784798

ABSTRACT

Vaccination in the developing world is hampered by limited patient access, which prevents individuals from receiving the multiple injections necessary for protective immunity. Here, we developed an injectable microparticle formulation of the inactivated polio vaccine (IPV) that releases multiple pulses of stable antigen over time. To accomplish this, we established an IPV stabilization strategy using cationic polymers for pH modulation to enhance traditional small-molecule-based stabilization methods. We investigated the mechanism of this strategy and showed that it was broadly applicable to all three antigens in IPV. Our lead formulations released two bursts of IPV 1 month apart, mimicking a typical vaccination schedule in the developing world. One injection of the controlled-release formulations elicited a similar or better neutralizing response in rats, considered the correlate of protection in humans, than multiple injections of liquid vaccine. This single-administration vaccine strategy has the potential to improve vaccine coverage in the developing world.


Subject(s)
Immunization Schedule , Poliovirus Vaccine, Inactivated/administration & dosage , Vaccination/methods , Animals , Disease Models, Animal , Female , Humans , Injections/methods , Microspheres , Poliomyelitis/prevention & control , Rats , Rats, Wistar
4.
Infect Immun ; 87(5)2019 03.
Article in English | MEDLINE | ID: mdl-30804099

ABSTRACT

Influenza kills 30,000 to 40,000 people each year in the United States and causes 10 times as many hospitalizations. A common complication of influenza is bacterial superinfection, which exacerbates morbidity and mortality from the viral illness. Recently, methicillin-resistant Staphylococcus aureus (MRSA) has emerged as the dominant pathogen found in bacterial superinfection, with Streptococcus pneumoniae a close second. However, clinicians have few tools to treat bacterial superinfection. Current therapy for influenza/bacterial superinfection consists of treating the underlying influenza infection and adding various antibiotics, which are increasingly rendered ineffective by rising bacterial multidrug resistance. Several groups have recently proposed the use of the antiviral cytokine interferon lambda (IFN-λ) as a therapeutic for influenza, as administration of pegylated IFN-λ improves lung function and survival during influenza by reducing the overabundance of neutrophils in the lung. However, our data suggest that therapeutic IFN-λ impairs bacterial clearance during influenza superinfection. Specifically, mice treated with an adenoviral vector to overexpress IFN-λ during influenza infection exhibited increased bacterial burdens upon superinfection with either MRSA or S. pneumoniae Surprisingly, adhesion molecule expression, antimicrobial peptide production, and reactive oxygen species activity were not altered by IFN-λ treatment. However, neutrophil uptake of MRSA and S. pneumoniae was significantly reduced upon IFN-λ treatment during influenza superinfection in vivo Together, these data support the theory that IFN-λ decreases neutrophil motility and function in the influenza-infected lung, which increases the bacterial burden during superinfection. Thus, we believe that caution should be exercised in the possible future use of IFN-λ as therapy for influenza.


Subject(s)
Antiviral Agents/therapeutic use , Influenza, Human/complications , Influenza, Human/drug therapy , Interferons/therapeutic use , Staphylococcus aureus/drug effects , Streptococcus pneumoniae/drug effects , Superinfection/drug therapy , Animals , Disease Models, Animal , Disease Susceptibility , Humans , Male , Mice , Mice, Inbred C57BL , Staphylococcal Infections/pathology , Superinfection/etiology , United States
5.
Biochem Biophys Res Commun ; 497(2): 700-704, 2018 03 04.
Article in English | MEDLINE | ID: mdl-29458021

ABSTRACT

We previously showed that the mitochondrial fatty acid oxidation enzyme long-chain acyl-CoA dehydrogenase (LCAD) is expressed in alveolar type II pneumocytes and that LCAD-/- mice have altered breathing mechanics and surfactant defects. Here, we hypothesized that LCAD-/- mice would be susceptible to influenza infection. Indeed, LCAD-/- mice demonstrated increased mortality following infection with 2009 pandemic influenza (A/CA/07/09). However, the mortality was not due to increased lung injury, as inflammatory cell counts, viral titers, and histology scores all showed non-significant trends toward milder injury in LCAD-/- mice. To confirm this, LCAD-/- were infected with a second, mouse-adapted H1N1 virus (A/PR/8/34), to which they responded with significantly less lung injury. While both strains become increasingly hypoglycemic over the first week post-infection, LCAD-/- mice lose body weight more rapidly than wild-type mice. Surprisingly, while acutely fasted LCAD-/- mice develop hepatic steatosis, influenza-infected LCAD-/- mice do not. They do, however, become more hypothermic than wild-type mice and demonstrate increased blood lactate values. We conclude that LCAD-/- mice succumb to influenza from bioenergetic starvation, likely due to increased reliance upon glucose for energy.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/genetics , Gene Knockdown Techniques , Influenza A Virus, H1N1 Subtype/physiology , Lung/pathology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , Animals , Body Weight , Female , Hypothermia/etiology , Hypothermia/genetics , Hypothermia/pathology , Hypothermia/virology , Lung/virology , Mice , Mice, Knockout , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/virology
6.
Am J Pathol ; 187(4): 851-863, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28193481

ABSTRACT

Seasonal and pandemic influenza is a cause of morbidity and mortality worldwide. Most people infected with influenza virus display mild-to-moderate disease phenotypes and recover within a few weeks. Influenza is known to cause persistent alveolitis in animal models; however, little is known about the molecular pathways involved in this phenotype. We challenged C57BL/6 mice with influenza A/PR/8/34 and examined lung pathologic processes and inflammation, as well as transcriptomic and epigenetic changes at 21 to 60 days after infection. Influenza induced persistent parenchymal lung inflammation, alveolar epithelial metaplasia, and epithelial endoplasmic reticulum stress that were evident after the clearance of virus and resolution of morbidity. Influenza infection induced robust changes in the lung transcriptome, including a significant impact on inflammatory and extracellular matrix protein expression. Despite the robust changes in lung gene expression, preceding influenza (21 days) did not exacerbate secondary Staphylococcus aureus infection. Finally, we examined the impact of influenza on miRNA expression in the lung and found an increase in miR-155. miR-155 knockout mice recovered from influenza infection faster than controls and had decreased lung inflammation and endoplasmic reticulum stress. These data illuminate the dynamic molecular changes in the lung in the weeks after influenza infection and characterize the repair process, identifying a novel role for miR-155.


Subject(s)
Epigenesis, Genetic , Lung/metabolism , Lung/virology , Orthomyxoviridae Infections/genetics , Transcriptome/genetics , Wound Healing/genetics , Animals , Disease Progression , Endoplasmic Reticulum Stress/genetics , Epithelium/pathology , Gene Expression Profiling , Inflammation/pathology , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Pneumonia/etiology , Pneumonia/microbiology , T-Lymphocytes/immunology , Time Factors
7.
J Immunol ; 196(3): 963-77, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26729801

ABSTRACT

IL-13 and IL-17A, produced mainly by Th2 and Th17 cells, respectively, have an influential role in asthma pathogenesis. We examined the role of IL-13 and IL-17A in mediating airway hyperresponsiveness (AHR), lung inflammation, and mucus metaplasia in a dual Th2/Th17 model of asthma. IL-13 and/or IL-17A were neutralized using mAbs. Th2/Th17 adoptive transfer induced a mixed asthma phenotype characterized by elevated eosinophilia and neutrophilia, tissue inflammation, mucus metaplasia, and AHR that were partially reversible with steroid treatment. Pulmonary inflammation and quasi-static lung compliance were largely unaffected by neutralization of IL-13 and/or IL-17A. However, neutralization of IL-13 alone or in combination with IL-17A significantly attenuated AHR and mucus metaplasia. Further, STAT6 activation was attenuated following IL-13 and IL-13/IL-17A Ab treatment. We next assessed the role of STAT6 in Th2/Th17-mediated allergic airway disease using STAT6(-/-) mice. STAT6(-/-) mice adoptively transferred with Th2/Th17 cells had decreased AHR compared with controls. These data suggest that IL-13 drives AHR and mucus metaplasia in a STAT6-dependent manner, without directly contributing to airway or tissue inflammation. IL-17A independently contributes to AHR, but it only partially mediates inflammation and mucus metaplasia in a mixed Th2/Th17 model of steroid-resistant asthma.


Subject(s)
Asthma/immunology , Interleukin-13/immunology , Interleukin-17/immunology , Respiratory Hypersensitivity/immunology , Adoptive Transfer , Animals , Asthma/pathology , Disease Models, Animal , Drug Resistance , Immunoblotting , Metaplasia/immunology , Metaplasia/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Mice, Transgenic , Mucus/immunology , Polymerase Chain Reaction , Respiratory Hypersensitivity/pathology , Th17 Cells/immunology , Th2 Cells/immunology , Transcriptome
8.
J Cell Mol Med ; 21(10): 2542-2552, 2017 10.
Article in English | MEDLINE | ID: mdl-28402065

ABSTRACT

The retinal pigment epithelium located between the neurosensory retina and the choroidal vasculature is critical for the function and maintenance of both the photoreceptors and underlying capillary endothelium. While the trophic role of retinal pigment epithelium on choroidal endothelial cells is well recognized, the existence of a reciprocal regulatory function of endothelial cells on retinal pigment epithelium cells remained to be fully characterized. Using a physiological long-term co-culture system, we determined the effect of retinal pigment epithelium-endothelial cell heterotypic interactions on cell survival, behaviour and matrix deposition. Human retinal pigment epithelium and endothelial cells were cultured on opposite sides of polyester transwells for up to 4 weeks in low serum conditions. Cell viability was quantified using a trypan blue assay. Cellular morphology was evaluated by H&E staining, S.E.M. and immunohistochemistry. Retinal pigment epithelium phagocytic function was examined using a fluorescent bead assay. Gene expression analysis was performed on both retinal pigment epithelium and endothelial cells by quantitative PCR. Quantification of extracellular matrix deposition was performed on decellularized transwells stained for collagen IV, fibronectin and fibrillin. Our results showed that presence of endothelial cells significantly improves retinal pigment epithelium maturation and function as indicated by the induction of visual cycle-associated genes, accumulation of a Bruch's membrane-like matrix and increase in retinal pigment epithelium phagocytic activity. Co-culture conditions led to increased expression of anti-angiogenic growth factors and receptors in both retinal pigment epithelium and endothelial cells compared to monoculture. Tube-formation assays confirmed that co-culture with retinal pigment epithelium significantly decreased the angiogenic phenotype of endothelial cells. These findings provide evidence of critical interdependent interactions between retinal pigment epithelium and endothelial cell involved in the maintenance of retinal homeostasis.


Subject(s)
Cell Communication , Coculture Techniques/methods , Human Umbilical Vein Endothelial Cells/cytology , Retinal Pigment Epithelium/cytology , Cell Line , Cell Survival , Cells, Cultured , Extracellular Matrix/metabolism , Gene Expression , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Phagocytosis , Retinal Pigment Epithelium/metabolism
9.
Am J Respir Crit Care Med ; 194(7): 807-820, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27007260

ABSTRACT

RATIONALE: Infection with Pneumocystis, an opportunistic fungal pathogen, can result in fulminant pneumonia in the clinical setting of patients with immunosuppression. In murine models, Pneumocystis has previously been shown to induce a CD4+ T cell-dependent eosinophilic response in the lung capable of providing protection. OBJECTIVES: We sought to explore the role of Pneumocystis in generating asthma-like lung pathology, given the natural eosinophilic response to infection. METHODS: Pneumocystis infection or antigen treatment was used to induce asthma-like pathology in wild-type mice. The roles of CD4+ T cells and eosinophils were examined using antibody depletion and knockout mice, respectively. The presence of anti-Pneumocystis antibodies in human serum samples was detected by ELISA and Western blotting. MEASUREMENTS AND MAIN RESULTS: Pneumocystis infection generates a strong type II response in the lung that requires CD4+ T cells. Pneumocystis infection was capable of priming a Th2 response similar to that of a commonly studied airway allergen, the house dust mite. Pneumocystis antigen treatment was also capable of inducing allergic inflammation in the lung, resulting in anti-Pneumocystis IgE production, goblet cell hyperplasia, and increased airway resistance. In the human population, patients with severe asthma had increased levels of anti-Pneumocystis IgG and IgE compared with healthy control subjects. Patients with severe asthma with elevated anti-Pneumocystis IgG levels had worsened symptom scores and lung parameters such as decreased forced expiratory volume and increased residual volume compared with patients with severe asthma who had low anti-Pneumocystis IgG. CONCLUSIONS: The present study demonstrates for the first time, to our knowledge, that Pneumocystis is an airway allergen capable of inducing asthma-like lung pathology.

10.
J Biol Chem ; 290(39): 23897-904, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26240137

ABSTRACT

The role of mitochondrial energy metabolism in maintaining lung function is not understood. We previously observed reduced lung function in mice lacking the fatty acid oxidation enzyme long-chain acyl-CoA dehydrogenase (LCAD). Here, we demonstrate that long-chain acylcarnitines, a class of lipids secreted by mitochondria when metabolism is inhibited, accumulate at the air-fluid interface in LCAD(-/-) lungs. Acylcarnitine accumulation is exacerbated by stress such as influenza infection or by dietary supplementation with l-carnitine. Long-chain acylcarnitines co-localize with pulmonary surfactant, a unique film of phospholipids and proteins that reduces surface tension and prevents alveolar collapse during breathing. In vitro, the long-chain species palmitoylcarnitine directly inhibits the surface adsorption of pulmonary surfactant as well as its ability to reduce surface tension. Treatment of LCAD(-/-) mice with mildronate, a drug that inhibits carnitine synthesis, eliminates acylcarnitines and improves lung function. Finally, acylcarnitines are detectable in normal human lavage fluid. Thus, long-chain acylcarnitines may represent a risk factor for lung injury in humans with dysfunctional fatty acid oxidation.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Carnitine/analogs & derivatives , Lung Injury/metabolism , Lung/metabolism , Phospholipids/metabolism , Pulmonary Surfactants/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Animals , Carnitine/genetics , Carnitine/metabolism , Humans , Lung/pathology , Lung Injury/genetics , Lung Injury/pathology , Mice , Mice, Knockout , Phospholipids/genetics
11.
J Biol Chem ; 289(15): 10668-10679, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24591516

ABSTRACT

Long-chain acyl-CoA dehydrogenase (LCAD) is a mitochondrial fatty acid oxidation enzyme whose expression in humans is low or absent in organs known to utilize fatty acids for energy such as heart, muscle, and liver. This study demonstrates localization of LCAD to human alveolar type II pneumocytes, which synthesize and secrete pulmonary surfactant. The physiological role of LCAD and the fatty acid oxidation pathway in lung was subsequently studied using LCAD knock-out mice. Lung fatty acid oxidation was reduced in LCAD(-/-) mice. LCAD(-/-) mice demonstrated reduced pulmonary compliance, but histological examination of lung tissue revealed no obvious signs of inflammation or pathology. The changes in lung mechanics were found to be due to pulmonary surfactant dysfunction. Large aggregate surfactant isolated from LCAD(-/-) mouse lavage fluid had significantly reduced phospholipid content as well as alterations in the acyl chain composition of phosphatidylcholine and phosphatidylglycerol. LCAD(-/-) surfactant demonstrated functional abnormalities when subjected to dynamic compression-expansion cycling on a constrained drop surfactometer. Serum albumin, which has been shown to degrade and inactivate pulmonary surfactant, was significantly increased in LCAD(-/-) lavage fluid, suggesting increased epithelial permeability. Finally, we identified two cases of sudden unexplained infant death where no lung LCAD antigen was detectable. Both infants were homozygous for an amino acid changing polymorphism (K333Q). These findings for the first time identify the fatty acid oxidation pathway and LCAD in particular as factors contributing to the pathophysiology of pulmonary disease.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Lipid Metabolism, Inborn Errors/metabolism , Lung Diseases/etiology , Pulmonary Surfactants/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Adult , Animals , Bronchi/metabolism , Cell Line, Tumor , Coenzyme A/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Fatty Acids/metabolism , Female , Homozygote , Humans , Infant , Infant, Newborn , Lung/metabolism , Lung Diseases/metabolism , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxygen/metabolism , Phosphatidylcholines/chemistry , Phosphatidylglycerols/chemistry , Polymorphism, Genetic , Pulmonary Alveoli/metabolism
12.
Am J Physiol Lung Cell Mol Physiol ; 309(2): L158-67, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26001778

ABSTRACT

Suppression of type 17 immunity by type I interferon (IFN) during influenza A infection has been shown to enhance susceptibility to secondary bacterial pneumonia. Although this mechanism has been described in coinfection with gram-positive bacteria, it is unclear whether similar mechanisms may impair lung defense against gram-negative infections. Furthermore, precise delineation of the duration of type I IFN-associated susceptibility to bacterial infection remains underexplored. Therefore, we investigated the effects of preceding influenza A virus infection on subsequent challenge with the gram-negative bacteria Escherichia coli or Pseudomonas aeruginosa and the temporal association between IFN expression with susceptibility to Staphylococcus aureus challenge in a mouse model of influenza and bacterial coinfection. Here we demonstrate that preceding influenza A virus led to increased lung E. coli and P. aeruginosa bacterial burden, which was associated with suppression of type 17 immunity and attenuation of antimicrobial peptide expression. Enhanced susceptibility to S. aureus coinfection ceased at day 14 of influenza infection, when influenza-associated type I IFN levels had returned to baseline levels, further suggesting a key role for type I IFN in coinfection pathogenesis. These findings further implicate type I IFN-associated suppression of type 17 immunity and antimicrobial peptide production as a conserved mechanism for enhanced susceptibility to both gram-positive and gram-negative bacterial coinfection during influenza infection.


Subject(s)
Escherichia coli Infections/microbiology , Influenza A virus/pathogenicity , Orthomyxoviridae Infections/microbiology , Pneumonia, Bacterial/microbiology , Pneumonia/microbiology , Receptor, Interferon alpha-beta/physiology , Staphylococcal Infections/microbiology , Animals , Antimicrobial Cationic Peptides/metabolism , Coinfection/immunology , Coinfection/microbiology , Coinfection/virology , Disease Susceptibility , Escherichia coli/immunology , Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Escherichia coli Infections/virology , Influenza A virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Pneumonia/immunology , Pneumonia/virology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/virology , Staphylococcal Infections/immunology , Staphylococcal Infections/virology , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity
13.
J Immunol ; 191(10): 5153-9, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24089191

ABSTRACT

Pneumonia is a leading cause of death worldwide. Staphylococcal aureus can be a cause of severe pneumonia alone or a common pathogen in secondary pneumonia following influenza. Recently, we reported that preceding influenza attenuated the Type 17 pathway, increasing the lung's susceptibility to secondary infection. IL-1ß is known to regulate host defense, including playing a role in Th17 polarization. We examined whether IL-1ß signaling is required for S. aureus host defense and whether influenza infection impacted S. aureus-induced IL-1ß production and subsequent Type 17 pathway activation. Mice were challenged with S. aureus (USA 300), with or without preceding Influenza A/PR/8/34 H1N1 infection. IL-1R1(-/-) mice had significantly higher S. aureus burden, increased mortality, and decreased Type 17 pathway activation following S. aureus challenge. Coinfected mice had significantly decreased IL-1ß production versus S. aureus infection alone at early time points following bacterial challenge. Preceding influenza did not attenuate S. aureus-induced inflammasome activation, but there was early suppression of NF-κB activation, suggesting an inhibition of NF-κB-dependent transcription of pro-IL-1ß. Furthermore, overexpression of IL-1ß in influenza and S. aureus-coinfected mice rescued the induction of IL-17 and IL-22 by S. aureus and improved bacterial clearance. Finally, exogenous IL-1ß did not significantly rescue S. aureus host defense during coinfection in IL-17RA(-/-) mice or in mice in which IL-17 and IL-22 activity were blocked. These data reveal a novel mechanism by which Influenza A inhibits S. aureus-induced IL-1ß production, resulting in attenuation of Type 17 immunity and increased susceptibility to bacterial infection.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Interleukin-1beta/metabolism , Orthomyxoviridae Infections/immunology , Pneumonia, Staphylococcal/immunology , Staphylococcus aureus/immunology , Animals , Bacterial Load/genetics , Bacterial Load/immunology , Coinfection/immunology , Coinfection/microbiology , Coinfection/virology , Enzyme Activation/immunology , Inflammasomes/immunology , Interleukin-17/biosynthesis , Interleukin-1beta/biosynthesis , Interleukins/biosynthesis , Lung/immunology , Lung/microbiology , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Receptors, Interleukin-1 Type I/deficiency , Receptors, Interleukin-1 Type I/genetics , Signal Transduction , Interleukin-22
14.
J Infect Dis ; 209(6): 865-75, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24072844

ABSTRACT

Influenza A represents a significant cause of morbidity and mortality worldwide. Bacterial complications of influenza A confer the greatest risk to patients. TH17 pathway inhibition has been implicated as a mechanism by which influenza A alters bacterial host defense. Here we show that preceding influenza causes persistent Staphylococcus aureus infection and suppression of TH17 pathway activation in mice. Influenza does not inhibit S. aureus binding and uptake by phagocytic cells but instead attenuates S. aureus induced TH17 related antimicrobial peptides necessary for bacterial clearance in the lung. Importantly, exogenous lipocalin 2 rescued viral exacerbation of S. aureus infection and decreased free iron levels in the bronchoalveolar lavage from mice coinfected with S. aureus and influenza. These findings indicate a novel mechanism by which influenza A inhibits TH17 immunity and increases susceptibility to secondary bacterial pneumonia. Identification of new mechanisms in the pathogenesis of bacterial pneumonia could lead to future therapeutic targets.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Influenza A virus/immunology , Orthomyxoviridae Infections/microbiology , Pneumonia, Staphylococcal/microbiology , Staphylococcus aureus/immunology , Analysis of Variance , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/microbiology , Bronchoalveolar Lavage Fluid/virology , Coinfection/microbiology , Coinfection/virology , Host-Pathogen Interactions/immunology , Influenza A virus/pathogenicity , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Pneumonia, Staphylococcal/immunology , Pneumonia, Staphylococcal/virology , Staphylococcus aureus/pathogenicity , Th17 Cells
15.
Am J Pathol ; 182(4): 1286-96, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23490254

ABSTRACT

Influenza infection is widespread in the United States and the world. Despite low mortality rates due to infection, morbidity is common and little is known about the molecular events involved in recovery. Influenza infection results in persistent distal lung remodeling, and the mechanism(s) involved are poorly understood. Recently IL-22 has been found to mediate epithelial repair. We propose that IL-22 is critical for recovery of normal lung function and architecture after influenza infection. Wild-type and IL-22(-/-) mice were infected with influenza A PR8/34 H1N1 and were followed up for up to 21 days post infection. IL-22 receptor was localized to the airway epithelium in naive mice but was expressed at the sites of parenchymal lung remodeling induced by influenza infection. IL-22(-/-) mice displayed exacerbated lung injury compared with wild-type mice, which correlated with decreased lung function 21 days post infection. Epithelial metaplasia was observed in wild-type mice but was not evident in IL-22(-/-) animals that were characterized with an increased fibrotic phenotype. Gene expression analysis revealed aberrant expression of epithelial genes involved in repair processes, among changes in several other biological processes. These data indicate that IL-22 is required for normal lung repair after influenza infection. IL-22 represents a novel pathway involved in interstitial lung disease.


Subject(s)
Epithelium/pathology , Epithelium/virology , Interleukins/metabolism , Lung/pathology , Lung/virology , Orthomyxoviridae Infections/pathology , Wound Healing , Animals , Basement Membrane/metabolism , Basement Membrane/pathology , Collagen/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Epithelium/metabolism , Gene Expression Profiling , Gene Expression Regulation , Interleukins/deficiency , Lung/physiopathology , Metaplasia , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae Infections/virology , Receptors, Interleukin/metabolism , Respiratory Function Tests , Signal Transduction/genetics , Interleukin-22
16.
Am J Orthod Dentofacial Orthop ; 145(6): 787-98, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24880850

ABSTRACT

INTRODUCTION: In this study, we used liquid chromatography-mass spectrometry (LC-MS) to investigate the differences in the composition of gingival crevicular fluid between resorbing deciduous molars and nonresorbing permanent teeth. The main goal was to identify novel biomarkers associated with root resorption. METHODS: Eleven children (4 boys, 7 girls) in the mixed dentition were selected to participate in this split-mouth design study, in which a deciduous second molar with radiographic evidence of root resorption served as the experimental site, and the permanent first molar on the contralateral quadrant was the control site. Gingival crevicular fluid was collected using absorbing strips. A total of 22 samples (11 root resorption, 11 control) were each analyzed with 1-dimensional LC-MS. The remaining samples were then pooled across the 11 patients and analyzed by 2-dimensional LC-MS. The output files were converted to mascot generic format, which can be used to perform protein identification with conventional search engines. RESULTS: The 2-dimensional LC-MS protocol was able to identify 2789 and 2421 proteins in the control and resorption pooled samples, respectively. In this population, we detected significantly upregulated and downregulated proteins in the teeth with root resorption. Interestingly, many of these proteins are characteristically found in exosomes. CONCLUSIONS: We identified novel proteins that might prove to be useful biomarkers of root resorption, individually or as part of a panel.


Subject(s)
Gingival Crevicular Fluid/chemistry , Root Resorption/metabolism , Albumins/analysis , Biomarkers/analysis , Child , Chromatography, Liquid/methods , Dentition, Mixed , Female , Humans , Male , Molar/metabolism , Proteins/analysis , Tandem Mass Spectrometry/methods , Tooth Exfoliation/metabolism , Tooth, Deciduous/metabolism
17.
ACS Nano ; 18(29): 19038-19053, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38979966

ABSTRACT

Surgical intervention is the most common first-line treatment for severe traumatic brain injuries (TBIs) associated with high intracranial pressure, while the complexity of these surgical procedures often results in complications. Surgeons often struggle to comprehensively evaluate the TBI status, making it difficult to select the optimal intervention strategy. Here, we introduce a fluorescence imaging-based technology that uses high-quality silver indium selenide-based quantum dots (QDs) for integrated TBI diagnosis and surgical guidance. These engineered, poly(ethylene glycol)-capped QDs emit in the near-infrared region, are resistant to phagocytosis, and importantly, are ultrastable after the epitaxial growth of an aluminum-doped zinc sulfide shell in the aqueous phase that renders the QDs resistant to long-term light irradiation and complex physiological environments. We found that intravenous injection of QDs enabled both the precise diagnosis of TBI in a mouse model and, more importantly, the comprehensive evaluation of the TBI status before, during, and after an operation to distinguish intracranial from superficial hemorrhages, provide real-time monitoring of the secondary hemorrhage, and guide the decision making on the evacuation of intracranial hematomas. This QD-based diagnostic and monitoring system could ultimately complement existing clinical tools for treating TBI, which may help surgeons improve patient outcomes and avoid unnecessary procedures.


Subject(s)
Brain Injuries, Traumatic , Quantum Dots , Quantum Dots/chemistry , Brain Injuries, Traumatic/diagnostic imaging , Animals , Mice , Optical Imaging , Water/chemistry , Fluorescence , Indium/chemistry , Male , Polyethylene Glycols/chemistry
18.
Cardiovasc Res ; 120(6): 630-643, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38230606

ABSTRACT

AIMS: Human pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) provide a platform to identify and characterize factors that regulate the maturation of CMs. The transition from an immature foetal to an adult CM state entails coordinated regulation of the expression of genes involved in myofibril formation and oxidative phosphorylation (OXPHOS) among others. Lysine demethylase 5 (KDM5) specifically demethylates H3K4me1/2/3 and has emerged as potential regulators of expression of genes involved in cardiac development and mitochondrial function. The purpose of this study is to determine the role of KDM5 in iPSC-CM maturation. METHODS AND RESULTS: KDM5A, B, and C proteins were mainly expressed in the early post-natal stages, and their expressions were progressively downregulated in the post-natal CMs and were absent in adult hearts and CMs. In contrast, KDM5 proteins were persistently expressed in the iPSC-CMs up to 60 days after the induction of myogenic differentiation, consistent with the immaturity of these cells. Inhibition of KDM5 by KDM5-C70 -a pan-KDM5 inhibitor, induced differential expression of 2372 genes, including upregulation of genes involved in fatty acid oxidation (FAO), OXPHOS, and myogenesis in the iPSC-CMs. Likewise, genome-wide profiling of H3K4me3 binding sites by the cleavage under targets and release using nuclease assay showed enriched of the H3K4me3 peaks at the promoter regions of genes encoding FAO, OXPHOS, and sarcomere proteins. Consistent with the chromatin and gene expression data, KDM5 inhibition increased the expression of multiple sarcomere proteins and enhanced myofibrillar organization. Furthermore, inhibition of KDM5 increased H3K4me3 deposits at the promoter region of the ESRRA gene and increased its RNA and protein levels. Knockdown of ESRRA in KDM5-C70-treated iPSC-CM suppressed expression of a subset of the KDM5 targets. In conjunction with changes in gene expression, KDM5 inhibition increased oxygen consumption rate and contractility in iPSC-CMs. CONCLUSION: KDM5 inhibition enhances maturation of iPSC-CMs by epigenetically upregulating the expressions of OXPHOS, FAO, and sarcomere genes and enhancing myofibril organization and mitochondrial function.


Subject(s)
Cell Differentiation , Fatty Acids , Myocytes, Cardiac , Myofibrils , Oxidative Phosphorylation , Retinoblastoma-Binding Protein 2 , Humans , Cells, Cultured , Fatty Acids/metabolism , Gene Expression Regulation, Developmental , Histones/metabolism , Histones/genetics , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/enzymology , Mitochondria, Heart/enzymology , Mitochondria, Heart/metabolism , Mitochondria, Heart/genetics , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Myofibrils/enzymology , Oxidation-Reduction , Promoter Regions, Genetic , Retinoblastoma-Binding Protein 2/metabolism , Retinoblastoma-Binding Protein 2/genetics
19.
bioRxiv ; 2024 Mar 09.
Article in English | MEDLINE | ID: mdl-38496534

ABSTRACT

Toll-like receptors (TLRs) recognize pathogen- and damage-associated molecular patterns and, in turn, trigger the release of cytokines and other immunostimulatory molecules. As a result, TLR agonists are increasingly being investigated as vaccine adjuvants, though many of these agonists are small molecules that quickly diffuse away from the vaccination site, limiting their co-localization with antigens and, thus, their effect. Here, the small-molecule TLR7 agonist 1V209 is conjugated to a positively-charged multidomain peptide (MDP) hydrogel, K 2 , which was previously shown to act as an adjuvant promoting humoral immunity. Mixing the 1V209-conjugated K 2 50:50 with the unfunctionalized K 2 produces hydrogels that retain the shear-thinning and self-healing physical properties of the original MDP, while improving the solubility of 1V209 more than 200-fold compared to the unconjugated molecule. When co-delivered with ovalbumin as a model antigen, 1V209-functionalized K 2 produces antigen-specific IgG titers that were statistically similar to alum, the gold standard adjuvant, and a significantly lower ratio of Th2-associated IgG1 to Th1-associated IgG2a than alum, suggesting a more balanced Th1 and Th2 response. Together, these results suggest that K 2 MDP hydrogels functionalized with 1V209 are a promising adjuvant for vaccines against infectious diseases, especially those benefiting from a combined Th1 and Th2 immune response. Table of Contents: Activation of toll-like receptors (TLRs) stimulates a signaling cascade to induce an immune response. A TLR7 agonist was conjugated to an injectable peptide hydrogel, which was then used to deliver a model vaccine antigen. This platform produced antibody titers similar to the gold standard adjuvant alum and demonstrated an improved balance between Th1- and Th2-mediated immunity over alum.

20.
bioRxiv ; 2024 May 23.
Article in English | MEDLINE | ID: mdl-38826442

ABSTRACT

Maintaining safe and potent pharmaceutical drug levels is often challenging. Multidomain peptides (MDPs) assemble into supramolecular hydrogels with a well-defined, highly porous nanostructure that makes them attractive for drug delivery, yet their ability to extend release is typically limited by rapid drug diffusion. To overcome this challenge, we developed self-assembling boronate ester release (SABER) MDPs capable of engaging in dynamic covalent bonding with payloads containing boronic acids (BAs). As examples, we demonstrate that SABER hydrogels can prolong the release of five BA-containing small-molecule drugs as well as BA-modified insulin and antibodies. Pharmacokinetic studies revealed that SABER hydrogels extended the therapeutic effect of ganfeborole from days to weeks, preventing Mycobacterium tuberculosis growth better than repeated oral administration in an infection model. Similarly, SABER hydrogels extended insulin activity, maintaining normoglycemia for six days in diabetic mice after a single injection. These results suggest that SABER hydrogels present broad potential for clinical translation.

SELECTION OF CITATIONS
SEARCH DETAIL