Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Public Health ; 5: 59, 2017.
Article in English | MEDLINE | ID: mdl-28421176

ABSTRACT

BACKGROUND: Ethiopia has a significant paucity of available health-care workers. Despite the increasing number of medical schools, there are not enough physician instructors. Furthermore, availability and standardization of postgraduate training are lacking. Modalities of e-learning have been shown to be successful when used to impart medical education in other resource-limited countries. The Emory University and Addis Ababa University (AAU) Departments of Anesthesiology have formed a collaboration with the intent of improving the AAU Anesthesiology residency program, one of two postgraduate training programs for anesthesiology in Ethiopia. METHODS: An initial educational needs assessment identified areas in the existing training program that required improvement. In this pilot study, we describe how the current classroom-based curriculum is augmented by the introduction of interactive educational sessions and distributed learning in the form of video lectures. Video lectures covered topics based on areas identified by Ethiopian residents and faculty. Interactive sessions included hands-on ultrasound workshops and epidural placement practicums, a journal club, problem-based learning sessions, and a mock code simulation. Assessment of the additions of the newly introduced blended learning technique was conducted via pre- and posttests on the topics presented. RESULTS: Pre- to posttest score averages increased from 54.5% to 83.6%. CONCLUSION: An expansion of educational resources and modes of didactics are needed to fill the gaps that exist in Ethiopian anesthesiology training. Incorporating distributed learning into the existing didactic structure may lead to more efficacious instruction resulting in a higher retention rate of information.

2.
Am J Physiol Lung Cell Mol Physiol ; 300(6): L821-30, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21335520

ABSTRACT

Cigarette smoking is the major cause of chronic obstructive pulmonary disease (COPD) and predisposes subjects to severe respiratory tract infections. Epidemiological studies have shown that cigarette smokers are seven times more likely to contract influenza infection than nonsmokers. The mechanisms underlying this increased susceptibility are poorly characterized. Retinoic acid-inducible gene (RIG)-I is believed to play an important role in the recognition of, and response to, influenza virus and other RNA viruses. Our study focused on how cigarette smoke extract (CSE) alters the influenza-induced proinflammatory response and suppresses host antiviral activity in the human lung using a unique lung organ culture model. We first determined that treatment with 2-20% CSE did not induce cytotoxicity as assessed by LDH release. However, CSE treatment inhibited influenza-induced IFN-inducible protein 10 protein and mRNA expression. Induction of the major antiviral cytokine IFN-ß mRNA was also decreased by CSE. CSE also blunted viral-mediated RIG-I mRNA and protein expression. Inhibition of viral-mediated RIG-I induction by CSE was prevented by the antioxidants N-acetyl-cysteine and glutathione. These findings show that CSE suppresses antiviral and innate immune responses in influenza virus-infected human lungs through oxidative inhibition of viral-mediated induction of the pattern recognition receptor RIG-I. This immunosuppressive effect of CSE may play a role in the enhanced susceptibility of smokers to serious influenza infection in the lung.


Subject(s)
DEAD-box RNA Helicases/antagonists & inhibitors , Immunity, Innate/drug effects , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/metabolism , Lung/virology , Smoking/adverse effects , Antioxidants/pharmacology , Blotting, Western , Cytokines/metabolism , DEAD Box Protein 58 , DEAD-box RNA Helicases/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Immunoenzyme Techniques , Immunosuppression Therapy , Interferon-beta/metabolism , L-Lactate Dehydrogenase/metabolism , RNA, Messenger/genetics , Receptors, Immunologic , Reverse Transcriptase Polymerase Chain Reaction
3.
BMC Complement Altern Med ; 10: 69, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21078173

ABSTRACT

BACKGROUND: Influenza is a significant cause of morbidity and mortality. The recent pandemic of a novel H1N1 influenza virus has stressed the importance of the search for effective treatments for this disease. Essential oils from aromatic plants have been used for a wide variety of applications, such as personal hygiene, therapeutic massage and even medical practice. In this paper, we investigate the potential role of an essential oil in antiviral activity. METHODS: We studied a commercial essential oil blend, On Guard™, and evaluated its ability in modulating influenza virus, A/PR8/34 (PR8), infection in Madin-Darby canine kidney (MDCK) cells. Influenza virus was first incubated with the essential oil and infectivity in MDCK cells was quantified by fluorescent focus assay (FFA). In order to determine the mechanism of effects of essential oil in viral infection inhibition, we measured hemagglutination (HA) activity, binding and internalization of untreated and oil-treated virus in MDCK cells by flow cytometry and immunofluorescence microscopy. In addition, the effect of oil treatment on viral transcription and translation were assayed by relative end-point RT-PCR and western blot analysis. RESULTS: Influenza virus infectivity was suppressed by essential oil treatment in a dose-dependent manner; the number of nascent viral particles released from MDCK cells was reduced by 90% and by 40% when virus was treated with 1:4,000 and 1:6,000 dilutions of the oil, respectively. Oil treatment of the virus also decreased direct infection of the cells as the number of infected MDCK cells decreased by 90% and 45% when virus was treated with 1:2,000 and 1:3,000 dilutions of the oil, respectively. This was not due to a decrease in HA activity, as HA was preserved despite oil treatment. In addition, oil treatment did not affect virus binding or internalization in MDCK cells. These effects did not appear to be due to cytotoxicity of the oil as MDCK cell viability was only seen with concentrations of oil that were 2 to 6 times greater than the doses that inhibited viral infectivity. RT-PCR and western blotting demonstrated that oil treatment of the virus inhibited viral NP and NS1 protein, but not mRNA expression. CONCLUSIONS: An essential oil blend significantly attenuates influenza virus PR8 infectivity in vitro without affecting viral binding or cellular internalization in MDCK cells. Oil treated virus continued to express viral mRNAs but had minimal expression of viral proteins, suggesting that the antiviral effect may be due to inhibition of viral protein translation.


Subject(s)
Antiviral Agents/therapeutic use , Influenza A Virus, H1N1 Subtype/drug effects , Oils, Volatile/therapeutic use , Orthomyxoviridae Infections/prevention & control , Plant Extracts/therapeutic use , Viral Proteins/antagonists & inhibitors , Animals , Antiviral Agents/pharmacology , Cell Line , Dogs , Dose-Response Relationship, Drug , Hemagglutination/drug effects , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/prevention & control , Influenza, Human/virology , Kidney/cytology , Oils, Volatile/pharmacology , Orthomyxoviridae Infections/virology , Phytotherapy , Plant Extracts/pharmacology , RNA, Messenger/metabolism
4.
J Gen Virol ; 91(Pt 5): 1155-63, 2010 May.
Article in English | MEDLINE | ID: mdl-20071488

ABSTRACT

Adenovirus (Ad) type 7 can cause severe infection, including pneumonia, in military recruits and children. The initial inflammation is a neutrophilic interstitial infiltration with neutrophilic alveolitis. Subsequently, monocytes become evident and, finally, there is a predominantly lymphocytic infiltrate. We have established that Ad7 infection of epithelial cells stimulates release of the neutrophil chemotaxin interleukin (IL)-8, and have extended these studies to a human lung tissue model. Here, we studied cytokine responses to Ad7 in human alveolar macrophages (HAM) and our human lung tissue model. Both ELISA and RNase-protection assay (RPA) data demonstrated that, upon Ad7 infection, IP-10 and MIP-1alpha/beta are released from HAM. IP-10 and MIP-1alpha/beta protein levels were induced 2- and 3-fold, respectively, in HAM 24 h after Ad7 infection. We then investigated induction of specific cytokines in human lung tissue by RPA and ELISA. The results showed that IL-8 and IL-6 were induced 8 h after infection and, by 24 h, levels of IL-8, IL-6, MIP-1alpha/beta and MCP-1 were all increased. IP-10, a monocyte and lymphocyte chemokine, was also induced 30-fold, but only 24 h after infection. Immunohistochemistry staining confirmed that IL-8 was only released from the epithelial cells of lung slices and not from macrophages. IP-10 was secreted from both macrophages and epithelial cells. Moreover, full induction of IP-10 is likely to require participation and cooperation of both epithelial cells and macrophages in intact lung. Understanding the cytokine and chemokine induction during Ad7 infection may lead to novel ways to modulate the response to this pathogen.


Subject(s)
Adenoviridae Infections/immunology , Adenoviruses, Human/immunology , Cytokines/metabolism , Immunity, Innate , Lung/immunology , Pneumonia, Viral/immunology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology
5.
Virology ; 396(2): 178-88, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-19913271

ABSTRACT

We studied cytokine responses to influenza virus PR8 (H1N1) and Oklahoma/309/06 (OK/06, H3N2) in a novel human lung tissue model. Exposure of the model to influenza virus rapidly activated the mitogen-activated protein kinase signaling (MAPK) pathways ERK, p38 and JNK. In addition, RNase protection assay demonstrated the induction of several cytokine and chemokine mRNAs by virus. This finding was reflected at the translational level as IL-6, MCP-1, MIP-1 alpha/beta, IL-8 and IP-10 proteins were induced as determined by ELISA. Immunohistochemistry for IP-10 and MIP-1 alpha revealed that alveolar epithelial cells and macrophages were the source of these two cytokines. Taken together, both PR8 and OK/06 cause similar induction of cytokines in human lung, although OK/06 is less effective at inducing the chemokines MCP-1 and IL-8. This human organ culture model should thus provide a relevant platform to study the biological responses of human lung to influenza virus infection.


Subject(s)
Immunity, Innate/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/immunology , Lung/virology , Chemokines/biosynthesis , Cytokines/biosynthesis , Enzyme Induction/immunology , Humans , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Influenza, Human/virology , Lung/immunology , Mitogen-Activated Protein Kinase Kinases/biosynthesis , Organ Culture Techniques , Signal Transduction/immunology
6.
J Immunol ; 183(9): 5799-806, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19812208

ABSTRACT

The etiologic agent of inhalational anthrax, Bacillus anthracis, produces virulence toxins that are important in the disease pathogenesis. Current studies suggest that mouse and human macrophages are susceptible to immunosuppressive effects of one of the virulence toxins, lethal toxin (LT). Thus a paradigm has emerged that holds that the alveolar macrophage (AM) does not play a significant role in the innate immune response to B. anthracis or defend against the pathogen as it is disabled by LT. This is inconsistent with animal models and autopsy studies that show minimal disease at the alveolar surface. We examined whether AM are immunosuppressed by LT. We found that human AM were relatively resistant to LT-mediated innate immune cytokine suppression, MEK cleavage, and induction of apoptosis as compared with mouse RAW 264.7 macrophages. Mouse AM and murine bone marrow-derived macrophages were also relatively resistant to LT-mediated apoptosis despite intermediate sensitivity to MEK cleavage. The binding component of LT, protective Ag, does not attach to human AM, although it did bind to mouse AM, murine bone marrow-derived macrophages, and RAW 264.7 macrophages. Human AM do not produce significant amounts of the protective Ag receptor anthrax toxin receptor 1 (TEM8/ANTXR1) and anthrax toxin receptor 2 (CMG2/ANTXR2). Thus, mature and differentiated AM are relatively resistant to the effects of LT as compared with mouse RAW 264.7 macrophages. AM resistance to LT may enhance clearance of the pathogen from the alveolar surface and explain why this surface is relatively free of B. anthracis in animal models and autopsy studies.


Subject(s)
Anthrax/immunology , Anthrax/mortality , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Immunity, Innate , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Animals , Anthrax/microbiology , Antigens, Bacterial/toxicity , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacterial Toxins/toxicity , Cell Line , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Humans , Immunosuppressive Agents/immunology , Immunosuppressive Agents/toxicity , Macrophages, Alveolar/enzymology , Mice , Mice, Inbred C57BL , Virulence/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...