Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 13: 1228275, 2023.
Article in English | MEDLINE | ID: mdl-37692170

ABSTRACT

Since December 2019, the world has been facing viral pandemic called COVID-19 (Coronavirus disease 2019) caused by a new beta-coronavirus named severe acute respiratory syndrome coronavirus-2, or SARS-CoV-2. COVID-19 patients may present with a wide range of symptoms, from asymptomatic to requiring intensive care support. The severe form of COVID-19 is often marked by an altered immune response and cytokine storm. Advanced age, age-related and underlying diseases, including metabolic syndromes, appear to contribute to increased COVID-19 severity and mortality suggesting a role for mitochondria in disease pathogenesis. Furthermore, since the immune system is associated with mitochondria and its damage-related molecular patterns (mtDAMPs), the host mitochondrial system may play an important role during viral infections. Viruses have evolved to modulate the immune system and mitochondrial function for survival and proliferation, which in turn could lead to cellular stress and contribute to disease progression. Recent studies have focused on the possible roles of mitochondria in SARS-CoV-2 infection. It has been suggested that mitochondrial hijacking by SARS-CoV-2 could be a key factor in COVID-19 pathogenesis. In this review, we discuss the roles of mitochondria in viral infections including SARS-CoV-2 infection based on past and present knowledge. Paying attention to the role of mitochondria in SARS-CoV-2 infection will help to better understand the pathophysiology of COVID-19 and to achieve effective methods of prevention, diagnosis, and treatment.


Subject(s)
COVID-19 , Humans , SARS-CoV-2 , Mitochondria , Critical Care , Cytokine Release Syndrome
2.
Viruses ; 15(3)2023 02 23.
Article in English | MEDLINE | ID: mdl-36992320

ABSTRACT

The emergence and availability of closely related clinical isolates of SARS-CoV-2 offers a unique opportunity to identify novel nonsynonymous mutations that may impact phenotype. Global sequencing efforts show that SARS-CoV-2 variants have emerged and then been replaced since the beginning of the pandemic, yet we have limited information regarding the breadth of variant-specific host responses. Using primary cell cultures and the K18-hACE2 mouse, we investigated the replication, innate immune response, and pathology of closely related, clinical variants circulating during the first wave of the pandemic. Mathematical modeling of the lung viral replication of four clinical isolates showed a dichotomy between two B.1. isolates with significantly faster and slower infected cell clearance rates, respectively. While isolates induced several common immune host responses to infection, one B.1 isolate was unique in the promotion of eosinophil-associated proteins IL-5 and CCL11. Moreover, its mortality rate was significantly slower. Lung microscopic histopathology suggested further phenotypic divergence among the five isolates showing three distinct sets of phenotypes: (i) consolidation, alveolar hemorrhage, and inflammation, (ii) interstitial inflammation/septal thickening and peribronchiolar/perivascular lymphoid cells, and (iii) consolidation, alveolar involvement, and endothelial hypertrophy/margination. Together these findings show divergence in the phenotypic outcomes of these clinical isolates and reveal the potential importance of nonsynonymous mutations in nsp2 and ORF8.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Mice , SARS-CoV-2/genetics , Genotype , Phenotype , Inflammation , Mice, Transgenic , Disease Models, Animal , Lung
3.
Front Pharmacol ; 13: 1020133, 2022.
Article in English | MEDLINE | ID: mdl-36532717

ABSTRACT

Microbial communities form an important symbiotic ecosystem within humans and have direct effects on health and well-being. Numerous exogenous factors including airborne triggers, diet, and drugs impact these established, but fragile communities across the human lifespan. Crosstalk between the mucosal microbiota and the immune system as well as the gut-lung axis have direct correlations to immune bias that may promote chronic diseases like asthma. Asthma initiation and pathogenesis are multifaceted and complex with input from genetic, epigenetic, and environmental components. In this review, we summarize and discuss the role of the airway microbiome in asthma, and how the environment, diet and therapeutics impact this low biomass community of microorganisms. We also focus this review on the pediatric and Black populations as high-risk groups requiring special attention, emphasizing that the whole patient must be considered during treatment. Although new culture-independent techniques have been developed and are more accessible to researchers, the exact contribution the airway microbiome makes in asthma pathogenesis is not well understood. Understanding how the airway microbiome, as a living entity in the respiratory tract, participates in lung immunity during the development and progression of asthma may lead to critical new treatments for asthma, including population-targeted interventions, or even more effective administration of currently available therapeutics.

4.
Front Cell Dev Biol ; 10: 836755, 2022.
Article in English | MEDLINE | ID: mdl-35386204

ABSTRACT

Mitochondria are multifunctional organelles of which ultrastructure is tightly linked to cell physiology. Accumulating evidence shows that mitochondrial remodeling has an impact on immune responses, but our current understanding of the mitochondrial architecture, interactions, and morphological changes in immune cells, mainly in eosinophils, is still poorly known. Here, we applied transmission electron microscopy (TEM), single-cell imaging analysis, and electron tomography, a technique that provides three-dimensional (3D) views at high resolution, to investigate mitochondrial dynamics in mouse eosinophils developing in cultures as well as in the context of inflammatory diseases characterized by recruitment and activation of these cells (mouse models of asthma, H1N1 influenza A virus (IAV) infection, and schistosomiasis mansoni). First, quantitative analyses showed that the mitochondrial area decrease 70% during eosinophil development (from undifferentiated precursor cells to mature eosinophils). Mitophagy, a consistent process revealed by TEM in immature but not in mature eosinophils, is likely operating in mitochondrial clearance during eosinophilopoiesis. Events of mitochondria interaction (inter-organelle membrane contacts) were also detected and quantitated within developing eosinophils and included mitochondria-endoplasmic reticulum, mitochondria-mitochondria, and mitochondria-secretory granules, all of them significantly higher in numbers in immature compared to mature cells. Moreover, single-mitochondrion analyses revealed that as the eosinophil matures, mitochondria cristae significantly increase in number and reshape to lamellar morphology. Eosinophils did not change (asthma) or reduced (IAV and Schistosoma infections) their mitochondrial mass in response to inflammatory diseases. However, asthma and schistosomiasis, but not IAV infection, induced amplification of both cristae numbers and volume in individual mitochondria. Mitochondrial cristae remodeling occurred in all inflammatory conditions with the proportions of mitochondria containing only lamellar or tubular, or mixed cristae (an ultrastructural aspect seen just in tissue eosinophils) depending on the tissue/disease microenvironment. The ability of mitochondria to interact with granules, mainly mobilized ones, was remarkably captured by TEM in eosinophils participating in all inflammatory diseases. Altogether, we demonstrate that the processes of eosinophilopoiesis and inflammation-induced activation interfere with the mitochondrial dynamics within mouse eosinophils leading to cristae remodeling and inter-organelle contacts. The understanding of how mitochondrial dynamics contribute to eosinophil immune functions is an open interesting field to be explored.

5.
Front Allergy ; 3: 852067, 2022.
Article in English | MEDLINE | ID: mdl-35386652

ABSTRACT

Asthma is a common chronic respiratory disease that affects millions of people worldwide. Patients with allergic asthma, the most prevalent asthma endotype, are widely considered to possess a defective immune response against some respiratory infectious agents, including viruses, bacteria and fungi. Furthermore, respiratory pathogens are associated with asthma development and exacerbations. However, growing data suggest that the immune milieu in allergic asthma may be beneficial during certain respiratory infections. Immunomodulatory asthma treatments, although beneficial, should then be carefully prescribed to avoid misuse and overuse as they can also alter the host microbiome. In this review, we summarize and discuss recent evidence of the correlations between allergic asthma and the most significant respiratory infectious agents that have a role in asthma pathogenesis. We also discuss the implications of current asthma therapeutics beyond symptom prevention.

6.
Cells ; 10(4)2021 04 15.
Article in English | MEDLINE | ID: mdl-33921169

ABSTRACT

Fungi represent one of the most diverse and abundant eukaryotes on earth, and their ubiquity and small proteolytically active products make them pervasive allergens that affect humans and other mammals. The immunologic parameters surrounding fungal allergies are still not fully elucidated despite their importance given that a large proportion of severe asthmatics are sensitized to fungal allergens. Herein, we explore fungal allergic asthma with emphasis on mouse models that recapitulate the characteristics of human disease, and the main leukocyte players in the pathogenesis of fungal allergies. The endogenous mycobiome may also contribute to fungal asthma, a phenomenon that we discuss only superficially, as much remains to be discovered.


Subject(s)
Asthma/immunology , Asthma/microbiology , Fungi/physiology , Immunization , Animals , Asthma/physiopathology , B-Lymphocytes/immunology , Disease Models, Animal , Humans , T-Lymphocytes/immunology
7.
Cells ; 10(3)2021 02 27.
Article in English | MEDLINE | ID: mdl-33673645

ABSTRACT

Eosinophils, previously considered terminally differentiated effector cells, have multifaceted functions in tissues. We previously found that allergic mice with eosinophil-rich inflammation were protected from severe influenza and discovered specialized antiviral effector functions for eosinophils including promoting cellular immunity during influenza. In this study, we hypothesized that eosinophil responses during the early phase of influenza contribute to host protection. Using in vitro and in vivo models, we found that eosinophils were rapidly and dynamically regulated upon influenza A virus (IAV) exposure to gain migratory capabilities to traffic to lymphoid organs after pulmonary infection. Eosinophils were capable of neutralizing virus upon contact and combinations of eosinophil granule proteins reduced virus infectivity through hemagglutinin inactivation. Bi-directional crosstalk between IAV-exposed epithelial cells and eosinophils occurred after IAV infection and cross-regulation promoted barrier responses to improve antiviral defenses in airway epithelial cells. Direct interactions between eosinophils and airway epithelial cells after IAV infection prevented virus-induced cytopathology in airway epithelial cells in vitro, and eosinophil recipient IAV-infected mice also maintained normal airway epithelial cell morphology. Our data suggest that eosinophils are important in the early phase of IAV infection providing immediate protection to the epithelial barrier until adaptive immune responses are deployed during influenza.


Subject(s)
Cell Adhesion Molecules/metabolism , Eosinophils/metabolism , Epithelial Cells/metabolism , Gene Expression Profiling/methods , Influenza A virus/genetics , Animals , Disease Models, Animal , Humans , Mice
8.
Methods Mol Biol ; 2241: 99-112, 2021.
Article in English | MEDLINE | ID: mdl-33486731

ABSTRACT

Eosinophils are granulocytes that were historically considered to be terminally differentiated at the time of bone marrow egress. However, more recent evidence provides a new outlook on these cells as complex immunomodulators that are involved in host defense and homeostasis. Our work established a role for eosinophils as mediators of antiviral immune responses during influenza in hosts that were sensitized and challenged with fungal allergens. Herein, we describe methods for working with murine eosinophils in the context of influenza A virus.


Subject(s)
Eosinophils/cytology , Mycoses/immunology , Virus Diseases/immunology , Allergens , Animals , Asthma/immunology , Disease Models, Animal , Dogs , Eosinophils/immunology , Influenza A virus/immunology , Influenza A virus/pathogenicity , Leukocyte Count , Madin Darby Canine Kidney Cells , Mice
9.
Biomedicines ; 8(10)2020 Oct 06.
Article in English | MEDLINE | ID: mdl-33036262

ABSTRACT

Asthma affects over 8% of the pediatric population in the United States, and Memphis, Tennessee has been labeled an asthma capital. Plasma samples were analyzed for biomarker profiles from 95 children with severe asthma and 47 age-matched, hospitalized nonasthmatic controls at Le Bonheur Children's Hospital in Memphis, where over 4000 asthmatics are cared for annually. Asthmatics exhibited significantly higher levels of periostin, surfactant protein D, receptor for advanced glycation end products and ß-hexosaminidase compared to controls. Children with severe asthma had lower levels of IgG1, IgG2 and IgA, and higher levels of IgE compared to controls, and approximately half of asthmatics exhibited IgG1 levels that were below age-specific norms. Vitamin A levels, measured by the surrogate retinol-binding protein, were insufficient or deficient in most asthmatic children, and correlated positively with IgG1. Which came first, asthma status or low levels of vitamin A and immunoglobulins? It is likely that inflammatory disease and immunosuppressive drugs contributed to a reduction in vitamin A and immunoglobulin levels. However, a nonmutually exclusive hypothesis is that low dietary vitamin A caused reductions in immune function and rendered children vulnerable to respiratory disease and consequent asthma pathogenesis. Continued attention to nutrition in combination with the biomarker profile is recommended to prevent and treat asthma in vulnerable children.

10.
J Leukoc Biol ; 108(1): 151-168, 2020 07.
Article in English | MEDLINE | ID: mdl-32386457

ABSTRACT

Allergic asthma and influenza are common respiratory diseases with a high probability of co-occurrence. During the 2009 influenza pandemic, hospitalized patients with influenza experienced lower morbidity if asthma was an underlying condition. We have previously demonstrated that acute allergic asthma protects mice from severe influenza and have implicated eosinophils in the airways of mice with allergic asthma as participants in the antiviral response. However, very little is known about how eosinophils respond to direct exposure to influenza A virus (IAV) or the microenvironment in which the viral burden is high. We hypothesized that eosinophils would dynamically respond to the presence of IAV through phenotypic, transcriptomic, and physiologic changes. Using our mouse model of acute fungal asthma and influenza, we showed that eosinophils in lymphoid tissues were responsive to IAV infection in the lungs and altered surface expression of various markers necessary for cell activation in a niche-specific manner. Siglec-F expression was altered in a subset of eosinophils after virus exposure, and those expressing high Siglec-F were more active (IL-5Rαhi CD62Llo ). While eosinophils exposed to IAV decreased their overall transcriptional activity and mitochondrial oxygen consumption, transcription of genes encoding viral recognition proteins, Ddx58 (RIG-I), Tlr3, and Ifih1 (MDA5), were up-regulated. CD8+ T cells from IAV-infected mice expanded in response to IAV PB1 peptide-pulsed eosinophils, and CpG methylation in the Tbx21 promoter was reduced in these T cells. These data offer insight into how eosinophils respond to IAV and help elucidate alternative mechanisms by which they regulate antiviral immune responses during IAV infection.


Subject(s)
Eosinophils/immunology , Influenza A virus/immunology , Animals , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, Viral/immunology , Asthma/immunology , Asthma/pathology , Asthma/virology , Bone Marrow Cells/pathology , CD8-Positive T-Lymphocytes/immunology , Cell Respiration/genetics , Chickens , DNA Demethylation , Dogs , Eosinophils/metabolism , Epigenesis, Genetic , Female , Interleukin-5 Receptor alpha Subunit/metabolism , Lung/pathology , Lung/virology , Madin Darby Canine Kidney Cells , Mice, Inbred C57BL , Mitochondria/metabolism , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Phenotype , Sialic Acid Binding Immunoglobulin-like Lectins , T-Box Domain Proteins/metabolism , Transcriptome/genetics , Up-Regulation
11.
Front Immunol ; 11: 3, 2020.
Article in English | MEDLINE | ID: mdl-32117216

ABSTRACT

The primary function of the respiratory system of gas exchange renders it vulnerable to environmental pathogens that circulate in the air. Physical and cellular barriers of the respiratory tract mucosal surface utilize a variety of strategies to obstruct microbe entry. Physical barrier defenses including the surface fluid replete with antimicrobials, neutralizing immunoglobulins, mucus, and the epithelial cell layer with rapidly beating cilia form a near impenetrable wall that separates the external environment from the internal soft tissue of the host. Resident leukocytes, primarily of the innate immune branch, also maintain airway integrity by constant surveillance and the maintenance of homeostasis through the release of cytokines and growth factors. Unfortunately, pathogens such as influenza virus and Streptococcus pneumoniae require hosts for their replication and dissemination, and prey on the respiratory tract as an ideal environment causing severe damage to the host during their invasion. In this review, we outline the host-pathogen interactions during influenza and post-influenza bacterial pneumonia with a focus on inter- and intra-cellular crosstalk important in pulmonary immune responses.


Subject(s)
Host-Pathogen Interactions/immunology , Influenza A virus/immunology , Influenza, Human/immunology , Pneumococcal Infections/immunology , Respiratory Mucosa/immunology , Streptococcus pneumoniae/immunology , Animals , Coinfection/immunology , Cytokines/metabolism , Epithelial Cells/immunology , Humans , Immunity, Innate , Influenza, Human/drug therapy , Influenza, Human/virology , Leukocytes/immunology , Lung/immunology , Mice , Pneumococcal Infections/drug therapy , Pneumococcal Infections/microbiology
12.
Sci Rep ; 9(1): 19360, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31852944

ABSTRACT

Asthma is a chronic airways condition that can be exacerbated during respiratory infections. Our previous work, together with epidemiologic findings that asthmatics were less likely to suffer from severe influenza during the 2009 pandemic, suggest that additional complications of influenza such as increased susceptibility to bacterial superinfection, may be mitigated in allergic hosts. To test this hypothesis, we developed a murine model of 'triple-disease' in which mice rendered allergic to Aspergillus fumigatus were co-infected with influenza A virus and Streptococcus pneumoniae seven days apart. Significant alterations to known synergistic effects of co-infection were noted in the allergic mice including reduced morbidity and mortality, bacterial burden, maintenance of alveolar macrophages, and reduced lung inflammation and damage. The lung microbiome of allergic mice differed from that of non-allergic mice during co-infection and antibiotic-induced perturbation to the microbiome rendered allergic animals susceptible to severe morbidity. Our data suggest that responses to co-infection in allergic hosts likely depends on the immune and microbiome states and that antibiotics should be used with caution in individuals with underlying chronic lung disease.


Subject(s)
Coinfection/microbiology , Coinfection/virology , Hypersensitivity/microbiology , Inflammation/microbiology , Influenza A Virus, H1N1 Subtype/physiology , Lung/microbiology , Microbiota , Streptococcus pneumoniae/physiology , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biodiversity , Coinfection/drug therapy , Coinfection/immunology , Hypersensitivity/complications , Hypersensitivity/drug therapy , Hypersensitivity/immunology , Inflammation/complications , Inflammation/drug therapy , Inflammation/immunology , Lung/drug effects , Lung/pathology , Lung/virology , Mice, Inbred C57BL , Microbiota/drug effects , Models, Biological , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/prevention & control , Pneumonia, Bacterial/complications , Pneumonia, Bacterial/drug therapy , Pneumonia, Bacterial/prevention & control
13.
J Immunol Res ; 2019: 8028725, 2019.
Article in English | MEDLINE | ID: mdl-31612153

ABSTRACT

Influenza virus infection is a serious threat to humans and animals, with the potential to cause severe pneumonia and death. Annual vaccination strategies are a mainstay to prevent complications related to influenza. However, protection from the emerging subtypes of influenza A viruses (IAV) even in vaccinated individuals is challenging. Innate immune cells are the first cells to respond to IAV infection in the respiratory tract. Virus replication-induced production of cytokines from airway epithelium recruits innate immune cells to the site of infection. These leukocytes, namely, neutrophils, monocytes, macrophages, dendritic cells, eosinophils, natural killer cells, innate lymphoid cells, and γδ T cells, become activated in response to IAV, to contain the virus and protect the airway epithelium while triggering the adaptive arm of the immune system. This review addresses different anti-influenza virus schemes of innate immune cells and how these cells fine-tune the balance between immunoprotection and immunopathology during IAV infection. Detailed understanding on how these innate responders execute anti-influenza activity will help to identify novel therapeutic targets to halt IAV replication and associated immunopathology.


Subject(s)
Influenza A virus/immunology , Influenza, Human/immunology , Leukocytes/virology , Cytokines , Dendritic Cells/immunology , Dendritic Cells/virology , Humans , Immunity, Innate , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Leukocytes/immunology , Macrophages/immunology , Macrophages/virology , Monocytes/immunology , Monocytes/virology , Virus Replication/immunology
14.
Curr Allergy Asthma Rep ; 19(8): 36, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31218528

ABSTRACT

PURPOSE OF REVIEW: Eosinophils are short-lived granulocytes that contain a variety of proteins and lipids traditionally associated with host defense against parasites. The primary goal of this review is to examine more recent evidence that challenged this rather outdated role of eosinophils in the context of pulmonary infections with helminths, viruses, and bacteria. RECENT FINDINGS: While eosinophil mechanisms that counter parasites, viruses, and bacteria are similar, the kinetics and impact may differ by pathogen type. Major antiparasitic responses include direct killing and immunoregulation, as well as some mechanisms by which parasite survival/growth is supported. Antiviral defenses may be as unembellished as granule protein-induced direct killing or more urbane as serving as a conduit for better adaptive immune responses to the invading virus. Although sacrificial, eosinophil DNA emitted in response to bacteria helps trap bacteria to limit dissemination. Herein, we discuss the current research redefining eosinophils as multifunctional cells that are active participants in host defense against lung pathogens. Eosinophils recognize and differentially respond to invading pathogens, allowing them to deploy innate defense mechanisms to contain and clear the infection, or modulate the immune response. Modern technology and animal models have unraveled hitherto unknown capabilities of this surreptitious cell that indubitably has more functions awaiting discovery.


Subject(s)
Eosinophils/immunology , Respiratory Tract Infections/blood , Humans
15.
Front Immunol ; 10: 3058, 2019.
Article in English | MEDLINE | ID: mdl-32038616

ABSTRACT

The underlying pathologies of sickle cell disease and asthma share many characteristics in terms of respiratory inflammation. The principal mechanisms of pulmonary inflammation are largely distinct, but activation of common pathways downstream of the initial inflammatory triggers may lead to exacerbation of both disease states. The altered inflammatory landscape of these respiratory pathologies can differentially impact respiratory pathogen susceptibility in patients with sickle cell disease and asthma. How these two distinct diseases behave in a comorbid setting can further exacerbate pulmonary complications associated with both disease states and impact susceptibility to respiratory infection. This review will provide a concise overview of how asthma distinctly affects individuals with sickle cell disease and how pulmonary physiology and inflammation are impacted during comorbidity.


Subject(s)
Anemia, Sickle Cell/immunology , Asthma/immunology , Hypersensitivity/immunology , Pneumonia/immunology , Respiratory Tract Infections/immunology , Humans , Signal Transduction
16.
Microb Pathog ; 127: 212-219, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30529429

ABSTRACT

Asthma and influenza are leading causes of worldwide morbidity and mortality. Although these two conditions can co-exist in the same patient, the immune parameters that impact disease outcomes are not fully elucidated. The importance of macrophages to both conditions suggested a role for CD14, a co-receptor for endotoxin, as a regulatory mechanism for innate immune responses during asthma and influenza co-morbidity. Herein, we hypothesized that parameters of influenza morbidity will be reduced in the absence of CD14. Age and gender matched wild-type (WT) and CD14 knock-out (KO) mice were subjected to our validated model of Aspergillus-induced model of asthma and/or influenza. Characteristics of disease pathogenesis were investigated using standard methods in weight loss, flow cytometry, airway resistance, histology, quantitative real-time PCR, and viral titer quantification. The absence of CD14 did not have an impact on morbidity as these mice were equally susceptible to disease with similar airway resistance. Peribronchovascular inflammation and goblet cell content were equivalent between WT and KO mice in asthma alone and asthma and influenza co-morbidity. Co-morbid KO mice had less lymphocytes and eosinophils in the airways although their lung viral burden was equivalent to WT. Inflammatory gene signatures were altered in co-morbid mice in each genotype. CD14 expression on macrophages is necessary for airway inflammation but not for viral pathogenesis in allergic hosts.


Subject(s)
Asthma/pathology , Lipopolysaccharide Receptors/metabolism , Macrophages/immunology , Macrophages/virology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae/immunology , Animals , Body Weight , Disease Models, Animal , Flow Cytometry , Histocytochemistry , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Viral Load
17.
Pharmaceutics ; 10(3)2018 Aug 01.
Article in English | MEDLINE | ID: mdl-30071698

ABSTRACT

Lung cancer (LC) is one of the leading causes of death in both men and women in the United States. Tannic acid (TA), a water-soluble polyphenol, exhibits a wide range of biological activities. TA has received much attention as a promising compound in the biomaterial and drug delivery fields. Lung fluid (LF) is a major barrier for distribution of drugs to the lungs. Therefore, the purpose of this study was to examine TA interaction with LF for effective delivery of anti-cancer drug molecules via pulmonary delivery. The extent of adsorption of LF proteins by TA was revealed by fluorescence quenching in fluorescence spectroscopy. The presence of LF in TA-LF complexes was noticed by the presence of protein peaks at 1653 cm-1. Both protein dot and SDS-PAGE analysis confirmed LF protein complexation at all TA concentrations employed. A stable particle TA-LF complex formation was observed through transmission electron microscopy (TEM) analysis. The complexation pattern measured by dynamic light scattering (DLS) indicated that it varies depending on the pH of the solutions. The degree of LF presence in TA-LF complexes signifies its interactive behavior in LC cell lines. Such superior interaction offered an enhanced anti-cancer activity of drugs encapsulated in TA-LF complex nanoformulations. Our results indicate that TA binds to LF and forms self-assemblies, which profoundly enhance interaction with LC cells. This study demonstrated that TA is a novel carrier for pharmaceutical drugs such as gemcitabine, carboplatin, and irinotecan.

18.
Front Immunol ; 9: 1843, 2018.
Article in English | MEDLINE | ID: mdl-30147697

ABSTRACT

Asthma and influenza are two pathologic conditions of the respiratory tract that affect millions worldwide. Influenza virus of the 2009 pandemic was highly transmissible and caused severe respiratory disease in young and middle-aged individuals. Asthma was discovered to be an underlying co-morbidity that led to hospitalizations during this influenza pandemic albeit with less severe outcomes. However, animal studies that investigated the relationship between allergic inflammation and pandemic (p)H1N1 infection, showed that while characteristics of allergic airways disease were exacerbated by this virus, governing immune responses that cause exacerbations may actually protect the host from severe outcomes associated with influenza. To better understand the relationship between asthma and severe influenza during the last pandemic, we conducted a systematic literature review of reports on hospitalized patients with asthma as a co-morbid condition during the pH1N1 season. Herein, we report that numerous other underlying conditions, such as cardiovascular, neurologic, and metabolic diseases may have been underplayed as major drivers of severe influenza during the 2009 pandemic. This review synopses, (1) asthma and influenza independently, (2) epidemiologic data surrounding asthma during the 2009 influenza pandemic, and (3) recent advances in our understanding of allergic host-pathogen interactions in the context of allergic airways disease and influenza in mouse models. Our goal is to showcase possible immunological benefits of allergic airways inflammation as countermeasures for influenza virus infections as a learning tool to discover novel pathways that can enhance our ability to hinder influenza virus replication and host pathology induced thereof.


Subject(s)
Asthma/immunology , Hypersensitivity/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Animals , Asthma/epidemiology , Comorbidity , Disease Models, Animal , Hospitalization , Host-Pathogen Interactions , Humans , Hypersensitivity/epidemiology , Inflammation , Influenza, Human/epidemiology , Mice , Orthomyxoviridae Infections/epidemiology , United States
19.
Sci Rep ; 8(1): 7061, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29728628

ABSTRACT

Asthma is characterized by inflammation and architectural changes in the lungs. A number of immune cells and mediators are recognized as initiators of asthma, although therapeutics based on these are not always effective. The multifaceted nature of this syndrome necessitate continued exploration of immunomodulators that may play a role in pathogenesis. We investigated the role of resistin-like molecule-beta (RELM-ß), a gut antibacterial, in the development and pathogenesis of Aspergillus-induced allergic airways disease. Age and gender matched C57BL/6J and Retnlb-/- mice rendered allergic to Aspergillus fumigatus were used to measure canonical markers of allergic asthma at early and late time points. Inflammatory cells in airways were similar, although Retnlb-/- mice had reduced tissue inflammation. The absence of RELM-ß elevated serum IgA and pro-inflammatory cytokines in the lungs at homeostasis. Markers of chronic disease including goblet cell numbers, Muc genes, airway wall remodelling, and hyperresponsiveness were greater in the absence RELM-ß. Specific inflammatory mediators important in antimicrobial defence in allergic asthma were also increased in the absence of RELM-ß. These data suggest that while characteristics of allergic asthma develop in the absence of RELM-ß, that RELM-ß may reduce the development of chronic markers of allergic airways disease.


Subject(s)
Asthma/diagnosis , Asthma/etiology , Disease Susceptibility , Hormones, Ectopic/genetics , Airway Remodeling/genetics , Airway Remodeling/immunology , Allergens/immunology , Animals , Asthma/metabolism , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Goblet Cells/metabolism , Immunity, Humoral , Immunoglobulin A/immunology , Inflammation Mediators/metabolism , Intercellular Signaling Peptides and Proteins , Lung/immunology , Lung/metabolism , Lung/pathology , Metaplasia , Mice , Mice, Knockout
20.
Biomed Res Int ; 2018: 3298378, 2018.
Article in English | MEDLINE | ID: mdl-29511677

ABSTRACT

Severe asthma with fungal sensitization predominates in the population suffering from allergic asthma, to which there is no cure. While corticosteroids are the mainstay in current treatment, other means of controlling inflammation may be beneficial. Herein, we hypothesized that mannan from Saccharomyces cerevisiae would dampen the characteristics of fungal allergic asthma by altering the pulmonary immune responses. Using wild-type and transgenic mice expressing the human mannose receptor on smooth muscle cells, we explored the outcome of mannan administration during allergen exposure on the pathogenesis of fungal asthma through measurement of cardinal features of disease such as inflammation, goblet cell number, and airway hyperresponsiveness. Mannan treatment did not alter most hallmarks of allergic airways disease in wild-type mice. Transgenic mice treated with mannan during allergen exposure had an equivalent response to non-mannan-treated allergic mice except for a prominent granulocytic influx into airways and cytokine availability. Our studies suggest no role for mannan as an inflammatory regulator during fungal allergy.


Subject(s)
Asthma/drug therapy , Hypersensitivity/drug therapy , Inflammation/drug therapy , Mannans/administration & dosage , Allergens/immunology , Allergens/toxicity , Animals , Aspergillus/immunology , Aspergillus/pathogenicity , Asthma/immunology , Asthma/microbiology , Asthma/pathology , Cytokines/genetics , Humans , Hypersensitivity/immunology , Hypersensitivity/microbiology , Hypersensitivity/pathology , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Inflammation/immunology , Inflammation/microbiology , Mannans/chemistry , Mice , Respiratory Hypersensitivity/drug therapy , Respiratory Hypersensitivity/immunology , Saccharomyces cerevisiae/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...