51
|
Aspergillus fumigatus Infection-Induced Neutrophil Recruitment and Location in the Conducting Airway of Immunocompetent, Neutropenic, and Immunosuppressed Mice. J Immunol Res 2018; 2018:5379085. [PMID: 29577051 PMCID: PMC5822902 DOI: 10.1155/2018/5379085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/05/2017] [Accepted: 11/22/2017] [Indexed: 12/27/2022] Open
Abstract
Susceptibility to fungal infection is commonly associated with impaired neutrophil responses. To study the mechanisms underlying this association, we investigated neutrophil recruitment to the conducting airway wall after Aspergillus fumigatus conidium inhalation in mouse models of drug-induced immunosuppression and antibody-mediated neutrophil depletion (neutropenia) by performing three-dimensional confocal laser-scanning microscopy of whole-mount primary bronchus specimens. Actin staining enabled visualization of the epithelial and smooth muscle layers that mark the airway wall. Gr-1+ or Ly6G+ neutrophils located between the epithelium and smooth muscles were considered airway wall neutrophils. The number of airway wall neutrophils for immunocompetent, immunosuppressed, and neutropenic mice before and 6 h after A. fumigatus infection were analyzed and compared. Our results show that the number of conducting airway wall neutrophils in immunocompetent mice significantly increased upon inflammation, while a dramatic reduction in this number was observed following immunosuppression and neutropenia. Interestingly, a slight increase in the infiltration of neutrophils into the airway wall was detected as a result of infection, even in immunosuppressed and neutropenic mice. Taken together, these data indicate that neutrophils are present in intact conducting airway walls and the number elevates upon A. fumigatus infection. Conducting airway wall neutrophils are affected by both neutropenia and immunosuppression.
Collapse
|
52
|
Cytosolic Phospholipase A 2α Promotes Pulmonary Inflammation and Systemic Disease during Streptococcus pneumoniae Infection. Infect Immun 2017; 85:IAI.00280-17. [PMID: 28808157 DOI: 10.1128/iai.00280-17] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/02/2017] [Indexed: 02/07/2023] Open
Abstract
Pulmonary infection by Streptococcus pneumoniae is characterized by a robust alveolar infiltration of neutrophils (polymorphonuclear cells [PMNs]) that can promote systemic spread of the infection if not resolved. We previously showed that 12-lipoxygenase (12-LOX), which is required to generate the PMN chemoattractant hepoxilin A3 (HXA3) from arachidonic acid (AA), promotes acute pulmonary inflammation and systemic infection after lung challenge with S. pneumoniae As phospholipase A2 (PLA2) promotes the release of AA, we investigated the role of PLA2 in local and systemic disease during S. pneumoniae infection. The group IVA cytosolic isoform of PLA2 (cPLA2α) was activated upon S. pneumoniae infection of cultured lung epithelial cells and was critical for AA release from membrane phospholipids. Pharmacological inhibition of this enzyme blocked S. pneumoniae-induced PMN transepithelial migration in vitro Genetic ablation of the cPLA2 isoform cPLA2α dramatically reduced lung inflammation in mice upon high-dose pulmonary challenge with S. pneumoniae The cPLA2α-deficient mice also suffered no bacteremia and survived a pulmonary challenge that was lethal to wild-type mice. Our data suggest that cPLA2α plays a crucial role in eliciting pulmonary inflammation during pneumococcal infection and is required for lethal systemic infection following S. pneumoniae lung challenge.
Collapse
|
53
|
Development of a Primary Human Co-Culture Model of Inflamed Airway Mucosa. Sci Rep 2017; 7:8182. [PMID: 28811631 PMCID: PMC5557980 DOI: 10.1038/s41598-017-08567-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/11/2017] [Indexed: 01/05/2023] Open
Abstract
Neutrophil breach of the mucosal surface is a common pathological consequence of infection. We present an advanced co-culture model to explore neutrophil transepithelial migration utilizing airway mucosal barriers differentiated from primary human airway basal cells and examined by advanced imaging. Human airway basal cells were differentiated and cultured at air-liquid interface (ALI) on the underside of 3 µm pore-sized transwells, compatible with the study of transmigrating neutrophils. Inverted ALIs exhibit beating cilia and mucus production, consistent with conventional ALIs, as visualized by micro-optical coherence tomography (µOCT). µOCT is a recently developed imaging modality with the capacity for real time two- and three-dimensional analysis of cellular events in marked detail, including neutrophil transmigratory dynamics. Further, the newly devised and imaged primary co-culture model recapitulates key molecular mechanisms that underlie bacteria-induced neutrophil transepithelial migration previously characterized using cell line-based models. Neutrophils respond to imposed chemotactic gradients, and migrate in response to Pseudomonas aeruginosa infection of primary ALI barriers through a hepoxilin A3-directed mechanism. This primary cell-based co-culture system combined with µOCT imaging offers significant opportunity to probe, in great detail, micro-anatomical and mechanistic features of bacteria-induced neutrophil transepithelial migration and other important immunological and physiological processes at the mucosal surface.
Collapse
|
54
|
Ghim M, Alpresa P, Yang SW, Braakman ST, Gray SG, Sherwin SJ, van Reeuwijk M, Weinberg PD. Visualization of three pathways for macromolecule transport across cultured endothelium and their modification by flow. Am J Physiol Heart Circ Physiol 2017; 313:H959-H973. [PMID: 28754719 DOI: 10.1152/ajpheart.00218.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/19/2017] [Accepted: 07/19/2017] [Indexed: 02/07/2023]
Abstract
Transport of macromolecules across vascular endothelium and its modification by fluid mechanical forces are important for normal tissue function and in the development of atherosclerosis. However, the routes by which macromolecules cross endothelium, the hemodynamic stresses that maintain endothelial physiology or trigger disease, and the dependence of transendothelial transport on hemodynamic stresses are controversial. We visualized pathways for macromolecule transport and determined the effect on these pathways of different types of flow. Endothelial monolayers were cultured under static conditions or on an orbital shaker producing different flow profiles in different parts of the wells. Fluorescent tracers that bound to the substrate after crossing the endothelium were used to identify transport pathways. Maps of tracer distribution were compared with numerical simulations of flow to determine effects of different shear stress metrics on permeability. Albumin-sized tracers dominantly crossed the cultured endothelium via junctions between neighboring cells, high-density lipoprotein-sized tracers crossed at tricellular junctions, and low-density lipoprotein-sized tracers crossed through cells. Cells aligned close to the angle that minimized shear stresses across their long axis. The rate of paracellular transport under flow correlated with the magnitude of these minimized transverse stresses, whereas transport across cells was uniformly reduced by all types of flow. These results contradict the long-standing two-pore theory of solute transport across microvessel walls and the consensus view that endothelial cells align with the mean shear vector. They suggest that endothelial cells minimize transverse shear, supporting its postulated proatherogenic role. Preliminary data show that similar tracer techniques are practicable in vivo.NEW & NOTEWORTHY Solutes of increasing size crossed cultured endothelium through intercellular junctions, through tricellular junctions, or transcellularly. Cells aligned to minimize the shear stress acting across their long axis. Paracellular transport correlated with the level of this minimized shear, but transcellular transport was reduced uniformly by flow regardless of the shear profile.
Collapse
Affiliation(s)
- Mean Ghim
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Paola Alpresa
- Department of Bioengineering, Imperial College London, London, United Kingdom.,Department of Aeronautics, Imperial College London, London, United Kingdom; and.,Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Sung-Wook Yang
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Sietse T Braakman
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Stephen G Gray
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Spencer J Sherwin
- Department of Aeronautics, Imperial College London, London, United Kingdom; and
| | - Maarten van Reeuwijk
- Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Peter D Weinberg
- Department of Bioengineering, Imperial College London, London, United Kingdom;
| |
Collapse
|
55
|
Abstract
Polymorphonuclear neutrophils (PMNs) are innate immune system cells that play an essential role in eradicating invading pathogens. PMN migration to sites of infection/inflammation requires exiting the microcirculation and subsequent crossing of epithelial barriers in mucosa-lined organs such as the lungs and intestines. Although these processes usually occur without significant damage to surrounding host tissues, dysregulated/excessive PMN transmigration and resultant bystander-tissue damage are characteristic of numerous mucosal inflammatory disorders. Mechanisms controlling PMN extravasation have been well characterized, but the molecular details regarding regulation of PMN migration across mucosal epithelia are poorly understood. Given that PMN migration across mucosal epithelia is strongly correlated with disease symptoms in many inflammatory mucosal disorders, enhanced understanding of the mechanisms regulating PMN transepithelial migration should provide insights into clinically relevant tissue-targeted therapies aimed at ameliorating PMN-mediated bystander-tissue damage. This review will highlight current understanding of the molecular interactions between PMNs and mucosal epithelia and the associated functional consequences.
Collapse
Affiliation(s)
- Jennifer C Brazil
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
56
|
Camp JV, Jonsson CB. A Role for Neutrophils in Viral Respiratory Disease. Front Immunol 2017; 8:550. [PMID: 28553293 PMCID: PMC5427094 DOI: 10.3389/fimmu.2017.00550] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 04/24/2017] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are immune cells that are well known to be present during many types of lung diseases associated with acute respiratory distress syndrome (ARDS) and may contribute to acute lung injury. Neutrophils are poorly studied with respect to viral infection, and specifically to respiratory viral disease. Influenza A virus (IAV) infection is the cause of a respiratory disease that poses a significant global public health concern. Influenza disease presents as a relatively mild and self-limiting although highly pathogenic forms exist. Neutrophils increase in the respiratory tract during infection with mild seasonal IAV, moderate and severe epidemic IAV infection, and emerging highly pathogenic avian influenza (HPAI). During severe influenza pneumonia and HPAI infection, the number of neutrophils in the lower respiratory tract is correlated with disease severity. Thus, comparative analyses of the relationship between IAV infection and neutrophils provide insights into the relative contribution of host and viral factors that contribute to disease severity. Herein, we review the contribution of neutrophils to IAV disease pathogenesis and to other respiratory virus infections.
Collapse
Affiliation(s)
- Jeremy V Camp
- Institute of Virology, University of Veterinary Medicine at Vienna, Vienna, Austria
| | - Colleen B Jonsson
- Department of Microbiology, University of Tennessee-Knoxville, Knoxville, TN, USA
| |
Collapse
|
57
|
Gomez JC, Doerschuk CM. Activating Integrins Isn't Always "Beta" for Neutrophil Migration! Am J Respir Cell Mol Biol 2017; 56:561-562. [PMID: 28459388 PMCID: PMC5449497 DOI: 10.1165/rcmb.2017-0066ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- John C Gomez
- 1 Marsico Lung Institute, Division of Pulmonary Diseases and Critical Care Medicine University of North Carolina Chapel Hill, North Carolina
| | - Claire M Doerschuk
- 1 Marsico Lung Institute, Division of Pulmonary Diseases and Critical Care Medicine University of North Carolina Chapel Hill, North Carolina
| |
Collapse
|
58
|
Illuminating dynamic neutrophil trans-epithelial migration with micro-optical coherence tomography. Sci Rep 2017; 8:45789. [PMID: 28368012 PMCID: PMC5377939 DOI: 10.1038/srep45789] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/02/2017] [Indexed: 01/24/2023] Open
Abstract
A model of neutrophil migration across epithelia is desirable to interrogate the underlying mechanisms of neutrophilic breach of mucosal barriers. A co-culture system consisting of a polarized mucosal epithelium and human neutrophils can provide a versatile model of trans-epithelial migration in vitro, but observations are typically limited to quantification of migrated neutrophils by myeloperoxidase correlation, a destructive assay that precludes direct longitudinal study. Our laboratory has recently developed a new isotropic 1-μm resolution optical imaging technique termed micro-optical coherence tomography (μOCT) that enables 4D (x,y,z,t) visualization of neutrophils in the co-culture environment. By applying μOCT to the trans-epithelial migration model, we can robustly monitor the spatial distribution as well as the quantity of neutrophils chemotactically crossing the epithelial boundary over time. Here, we demonstrate the imaging and quantitative migration results of our system as applied to neutrophils migrating across intestinal epithelia in response to a chemoattractant. We also demonstrate that perturbation of a key molecular event known to be critical for effective neutrophil trans-epithelial migration (CD18 engagement) substantially impacts this process both qualitatively and quantitatively.
Collapse
|
59
|
Escue R, Kandasamy K, Parthasarathi K. Thrombin Induces Inositol Trisphosphate-Mediated Spatially Extensive Responses in Lung Microvessels. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:921-935. [PMID: 28188112 DOI: 10.1016/j.ajpath.2016.12.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 12/09/2016] [Accepted: 12/20/2016] [Indexed: 12/20/2022]
Abstract
Activation of plasma membrane receptors initiates compartmentalized second messenger signaling. Whether this compartmentalization facilitates the preferential intercellular diffusion of specific second messengers is unclear. Toward this, the receptor-mediated agonist, thrombin, was instilled into microvessels in a restricted region of isolated blood-perfused mouse lungs. Subsequently, the thrombin-induced increase in endothelial F-actin was determined using confocal fluorescence microscopy. Increased F-actin was evident in microvessels directly treated with thrombin and in those located in adjoining thrombin-free regions. This increase was abrogated by inhibiting inositol trisphosphate-mediated calcium release with Xestospongin C (XeC). XeC also inhibited the thrombin-induced increase in the amplitude of endothelial cytosolic Ca2+ oscillations. Instillation of thrombin and XeC into adjacent restricted regions increased F-actin in microvessels in the thrombin-treated and adjacent regions but not in those in the XeC-treated region. Thus, inositol trisphosphate, and not calcium, diffused interendothelially to the spatially remote thrombin-free microvessels. Thus, activation of plasma membrane receptors increased the ambit of inflammatory responses via a second messenger different from that used by stimuli that induce cell-wide increases in second messengers. Thrombin however failed to induce the spatially extensive response in microvessels of mice lacking endothelial connexin43, suggesting a role for connexin43 gap junctions. Compartmental second messenger signaling and interendothelial communication define the specific second messenger involved in exacerbating proinflammatory responses to receptor-mediated agonists.
Collapse
Affiliation(s)
- Rachel Escue
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Kathirvel Kandasamy
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Kaushik Parthasarathi
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, Tennessee.
| |
Collapse
|
60
|
Hirano Y, Yang WL, Aziz M, Zhang F, Sherry B, Wang P. MFG-E8-derived peptide attenuates adhesion and migration of immune cells to endothelial cells. J Leukoc Biol 2017; 101:1201-1209. [PMID: 28096298 DOI: 10.1189/jlb.3a0416-184rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 12/20/2016] [Accepted: 12/28/2016] [Indexed: 02/05/2023] Open
Abstract
Milk fat globule-epidermal growth factor-factor 8 (MFG-E8) plays an immunomodulatory role in inflammatory diseases. MFG-E8-derived short peptide (MSP68) greatly reduces neutrophil infiltration and injury in the lung during sepsis. In this study, we examined the effect of MSP68 on chemotaxis of various immune cells and its regulatory mechanism. Bone marrow-derived neutrophils (BMDNs) from C57BL/6 mice, human monocyte THP-1 cell line, and human T lymphocyte Jurkat cell line were used for adhesion and migration assays using a Transwell method in the presence of MSP68. Treatment with MSP68 significantly inhibited the BMDN and THP-1 cell but not Jurkat cell adhesion on the TNF-α-stimulated pulmonary artery endothelial cell (PAEC) monolayer dose-dependently. MSP68 also significantly reduced BMDN adhesion on VCAM-1-coated wells dose dependently. Surface plasmon resonance (SPR) analysis revealed that MSP68 efficiently recognized integrin α4β1 (receptor for VCAM-1) at the dissociation constant (KD) of 1.53 × 10-7 M. These findings implicate that MSP68 prevents neutrophil adhesion to the activated endothelial cells by interfering with the binding between integrin α4β1 on neutrophils and VCAM-1 on endothelial cells. Moreover, MSP68 significantly attenuated the migration of BMDN and THP-1 cells but not Jurkat cells to their chemoattractants. Pretreatment with MSP68 inhibited the transmigration of BMDNs across the PAECs toward chemoattractants, fMLP, MIP-2, and complement fragment 5a (C5a) dose-dependently. Finally, we identified that the activation of p38 MAPK in BMDNs by fMLP was inhibited by MSP68. Thus, MSP68 attenuates extravasation of immune cells through the endothelial cell lining into inflamed tissue, implicating MSP68 to be a novel, therapeutic agent for inflammatory diseases caused by excessive immune cell infiltration.
Collapse
Affiliation(s)
- Yohei Hirano
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA.,Department of Emergency and Critical Care Medicine, Juntendo University and Urayasu Hospital, Chiba, Japan; and
| | - Weng-Lang Yang
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA.,Department of Surgery, Hofstra Northwell School of Medicine, Manhasset, New York, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Fangming Zhang
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Barbara Sherry
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA; .,Department of Surgery, Hofstra Northwell School of Medicine, Manhasset, New York, USA
| |
Collapse
|
61
|
Nguyen Thi Dieu T, Pham Nhat A, Craig TJ, Duong-Quy S. Clinical characteristics and cytokine changes in children with pneumonia requiring mechanical ventilation. J Int Med Res 2017; 45:1805-1817. [PMID: 28703632 PMCID: PMC5805188 DOI: 10.1177/0300060516672766] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective To assess clinical characteristics and cytokine levels in children with
severe pneumonia who required ventilatory support. Methods In this prospective, descriptive, cross-sectional study, blood and
endotracheal fluid samples were obtained from patients with severe
pneumonia, aged <5 years, within 24 h following intubation. Blood samples
were also obtained from age-matched healthy controls. Cytokine levels were
investigated using flow cytometry-assisted immunoassay. Results Forty-five patients with severe pneumonia requiring mechanical ventilation
(aged 10 ± 5 months) and 35 healthy age-matched controls were included.
Patients with severe pneumonia had significantly increased serum interleukin
(IL)-6, IL-8, and granulocyte/macrophage colony-stimulating factor
concentrations compared with controls (80.84 pg/ml versus 2.06 pg/ml,
90.03 pg/ml versus 6.62 pg/ml, and 115.58 pg/ml versus 11.47 pg/ml,
respectively). In the severe pneumonia group, serum IL-10 levels were
significantly higher in patients aged <6 months versus those aged 6–12
months. Age-group differences in serum cytokine levels did not correspond to
age-group differences in endotracheal-fluid cytokine levels. Serum IL-6
levels were significantly higher in patients who subsequently died versus
those who survived (267.12 pg/ml versus 20.75 pg/ml, respectively). Conclusion High IL-6 concentrations were associated with mortality in patients <5
years of age with severe pneumonia requiring mechanical ventilation.
Collapse
Affiliation(s)
- Thuy Nguyen Thi Dieu
- 1 Department of Immunology, Allergology and Rheumatology, National Hospital of Paediatrics, Hanoi Medical University, Hanoi, Vietnam
| | - An Pham Nhat
- 1 Department of Immunology, Allergology and Rheumatology, National Hospital of Paediatrics, Hanoi Medical University, Hanoi, Vietnam
| | - Timothy J Craig
- 2 Department of Pulmonary, Allergy and Critical Care Medicine, Penn State University, Hershey, PA, USA
| | - Sy Duong-Quy
- 2 Department of Pulmonary, Allergy and Critical Care Medicine, Penn State University, Hershey, PA, USA.,3 Department of Respiratory Diseases, Bio-Medical Research Centre, Lam Dong Medical College, Dalat, Vietnam.,4 Department of Respiratory and Lung Functional Exploration, Cochin Hospital, Paris Descartes University, Paris, France
| |
Collapse
|
62
|
Barth A, Brucker N, Moro AM, Nascimento S, Goethel G, Souto C, Fracasso R, Sauer E, Altknecht L, da Costa B, Duarte M, Menezes CB, Tasca T, Arbo MD, Garcia SC. Association between inflammation processes, DNA damage, and exposure to environmental pollutants. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2017; 24:353-362. [PMID: 27718115 DOI: 10.1007/s11356-016-7772-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 09/22/2016] [Indexed: 06/06/2023]
Abstract
Environmental exposure to pollutants, especially polycyclic aromatic hydrocarbons (PAHs), could lead to carcinogenesis development. However, there is a gap on the mechanisms involved in this effect. Therefore, the aim of this study was to investigate the potential relationship between exposure to environmental air pollution and inflammation process in DNA damage in taxi drivers. This study included 45 taxi drivers and 40 controls; non-smokers composed both groups. Biological monitoring was performed through quantification of urinary 1-hydroxypyrene (1-OHP). ICAM-1 (CD54) expression, NTPDase activity, inflammatory cytokine (IL-1β, IL-6, IL-10, TNF-α and IFN-γ) levels, and comet and micronucleus assays were evaluated. The results demonstrated that 1-OHP levels, ICAM-1 expression, NTPDase activity, and DNA damage biomarkers (% tail DNA and micronucleus frequency) were increased in taxi drivers compared to the control group (p < 0.01). Moreover, significant associations were found between 1-OHP levels and ICAM-1 expression, % tail DNA, and micronucleus frequency (p < 0.05). Besides, pro-inflammatory cytokine levels were positively correlated to % tail DNA and micronucleus frequency (p < 0.001). Our findings suggest an important association between environmental exposure to air pollution with increase of ICAM-1 expression and NTPDase activity in taxi drivers. Additionally, the multiple regression linear-analysis demonstrated association between IL-6 and DNA damage. Thus, the present study has provided important evidence that, in addition to environmental exposure to air pollutants, the inflammation process may contribute to DNA damage.
Collapse
Affiliation(s)
- Anelise Barth
- Laboratory of Toxicology (LATOX), Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, CEP.: 90610-000, Brazil
| | - Natália Brucker
- Laboratory of Toxicology (LATOX), Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, CEP.: 90610-000, Brazil
| | - Angela M Moro
- Laboratory of Toxicology (LATOX), Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, CEP.: 90610-000, Brazil
| | - Sabrina Nascimento
- Laboratory of Toxicology (LATOX), Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, CEP.: 90610-000, Brazil
- Post-graduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gabriela Goethel
- Laboratory of Toxicology (LATOX), Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, CEP.: 90610-000, Brazil
- Post-graduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Caroline Souto
- Laboratory of Toxicology (LATOX), Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, CEP.: 90610-000, Brazil
| | - Rafael Fracasso
- Laboratory of Toxicology (LATOX), Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, CEP.: 90610-000, Brazil
| | - Elisa Sauer
- Laboratory of Toxicology (LATOX), Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, CEP.: 90610-000, Brazil
- Post-graduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Louise Altknecht
- Laboratory of Toxicology (LATOX), Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, CEP.: 90610-000, Brazil
| | - Bárbara da Costa
- Laboratory of Toxicology (LATOX), Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, CEP.: 90610-000, Brazil
| | - Marta Duarte
- Department of Health Sciences, Lutheran University of Brazil, Santa Maria, RS, Brazil
| | - Camila B Menezes
- Post-graduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Laboratory of Research in Parasitology, Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Tiana Tasca
- Post-graduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Laboratory of Research in Parasitology, Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Marcelo D Arbo
- Laboratory of Toxicology (LATOX), Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, CEP.: 90610-000, Brazil
- Post-graduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Solange Cristina Garcia
- Laboratory of Toxicology (LATOX), Department of Analysis, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Avenida Ipiranga 2752, Santa Cecília, Porto Alegre, RS, CEP.: 90610-000, Brazil.
- Post-graduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Institute of Cardiology, University Cardiology Foundation, Porto Alegre, RS, Brazil.
| |
Collapse
|
63
|
Di Russo J, Hannocks MJ, Luik AL, Song J, Zhang X, Yousif L, Aspite G, Hallmann R, Sorokin L. Vascular laminins in physiology and pathology. Matrix Biol 2017; 57-58:140-148. [DOI: 10.1016/j.matbio.2016.06.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/28/2016] [Indexed: 12/11/2022]
|
64
|
Leukocyte Kinetics and Migration in the Lungs. Respir Med 2017. [DOI: 10.1007/978-3-319-41912-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
65
|
Abstract
Ischemic disorders, such as myocardial infarction, stroke, and peripheral vascular disease, are the most common causes of debilitating disease and death in westernized cultures. The extent of tissue injury relates directly to the extent of blood flow reduction and to the length of the ischemic period, which influence the levels to which cellular ATP and intracellular pH are reduced. By impairing ATPase-dependent ion transport, ischemia causes intracellular and mitochondrial calcium levels to increase (calcium overload). Cell volume regulatory mechanisms are also disrupted by the lack of ATP, which can induce lysis of organelle and plasma membranes. Reperfusion, although required to salvage oxygen-starved tissues, produces paradoxical tissue responses that fuel the production of reactive oxygen species (oxygen paradox), sequestration of proinflammatory immunocytes in ischemic tissues, endoplasmic reticulum stress, and development of postischemic capillary no-reflow, which amplify tissue injury. These pathologic events culminate in opening of mitochondrial permeability transition pores as a common end-effector of ischemia/reperfusion (I/R)-induced cell lysis and death. Emerging concepts include the influence of the intestinal microbiome, fetal programming, epigenetic changes, and microparticles in the pathogenesis of I/R. The overall goal of this review is to describe these and other mechanisms that contribute to I/R injury. Because so many different deleterious events participate in I/R, it is clear that therapeutic approaches will be effective only when multiple pathologic processes are targeted. In addition, the translational significance of I/R research will be enhanced by much wider use of animal models that incorporate the complicating effects of risk factors for cardiovascular disease. © 2017 American Physiological Society. Compr Physiol 7:113-170, 2017.
Collapse
Affiliation(s)
- Theodore Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Christopher P. Baines
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Ronald J. Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
66
|
Paul D, Baena V, Ge S, Jiang X, Jellison ER, Kiprono T, Agalliu D, Pachter JS. Appearance of claudin-5 + leukocytes in the central nervous system during neuroinflammation: a novel role for endothelial-derived extracellular vesicles. J Neuroinflammation 2016; 13:292. [PMID: 27852330 PMCID: PMC5112695 DOI: 10.1186/s12974-016-0755-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/31/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The mechanism of leukocyte transendothelial migration (TEM) across the highly restrictive blood-brain barrier (BBB) remains enigmatic, with paracellular TEM thought to require leukocytes to somehow navigate the obstructive endothelial tight junctions (TJs). Transient interactions between TJ proteins on the respective leukocyte and endothelial surfaces have been proposed as one mechanism for TEM. Given the expanding role of extracellular vesicles (EVs) in intercellular communication, we investigated whether EVs derived from brain microvascular endothelial cells (BMEC) of the BBB may play a role in transferring a major TJ protein, claudin-5 (CLN-5), to leukocytes as a possible basis for such a mechanism during neuroinflammation. METHODS High-resolution 3D confocal imaging was used to highlight CLN-5 immunoreactivity in the central nervous system (CNS) and on leukocytes of mice with the neuroinflammatory condition experimental autoimmune encephalomyelitis (EAE). Both Western blotting of circulating leukocytes from wild-type mice and fluorescence imaging of leukocyte-associated eGFP-CLN-5 in the blood and CNS of endothelial-targeted, Tie-2-eGFP-CLN-5 transgenic mice were used to confirm the presence of CLN-5 protein on these cells. EVs were isolated from TNF-α-stimulated BMEC cultures and blood plasma of Tie-2-eGFP-CLN-5 mice with EAE and evaluated for CLN-5 protein by Western blotting and fluorescence-activated cell sorting (FACS), respectively. Confocal imaging and FACS were used to detect binding of endothelial-derived EVs from these two sources to leukocytes in vitro. Serial electron microscopy (serial EM) and 3D contour-based surface reconstruction were employed to view EV-like structures at the leukocyte:BBB interface in situ in inflamed CNS microvessels. RESULTS A subpopulation of leukocytes immunoreactive for CLN-5 on their surface was seen to infiltrate the CNS of mice with EAE and reside in close apposition to inflamed vessels. Confocal imaging of immunostained samples and Western blotting established the presence of CLN-5+ leukocytes in blood as well, implying these cells are present prior to TEM. Moreover, imaging of inflamed CNS vessels and the associated perivascular cell infiltrates from Tie-2-eGFP-CLN-5 mice with EAE revealed leukocytes bearing the eGFP label, further supporting the hypothesis CLN-5 is transferred from endothelial cells to circulating leukocytes in vivo. Western blotting of BMEC-derived EVs, corresponding in size to both exosomes and microvesicles, and FACS analysis of plasma-derived EVs from Tie-2-eGFP-CLN-5 mice with EAE validated expression of CLN-5 by EVs of endothelial origin. Confocal imaging and FACS further revealed both PKH-67-labeled EVs from cultured BMECs and eGFP-CLN-5+ EVs from plasma of Tie-2-eGFP-CLN-5 mice with EAE can bind to leukocytes. Lastly, serial EM and 3D contour-based surface reconstruction revealed a close association of EV-like structures between the marginating leukocytes and BMECs in situ during EAE. CONCLUSIONS During neuroinflammation, CLN-5+ leukocytes appear in the CNS, and both CLN-5+ leukocytes and CLN-5+ EVs are detected in the blood. As endothelial cells transfer CLN-5+ to leukocytes in vivo, and EVs released from BMEC bind to leukocytes in vitro, EVs may serve as the vehicles to transfer CLN-5 protein at sites of leukocyte:endothelial contact along the BBB. This action may be a prelude to facilitate TEM through the formation of temporary TJ protein bridges between these two cell types.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Central Nervous System/diagnostic imaging
- Central Nervous System/pathology
- Cytokines/metabolism
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/blood
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Endothelial Cells/pathology
- Endothelial Cells/ultrastructure
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/ultrastructure
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/ultrastructure
- Female
- Leukocytes/metabolism
- Lysosomal Membrane Proteins
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Myelin-Oligodendrocyte Glycoprotein/immunology
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Peptide Fragments/immunology
- Peptide Fragments/toxicity
Collapse
Affiliation(s)
- Debayon Paul
- Blood-Brain Barrier Laboratory, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06070 USA
- Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06070 USA
| | - Valentina Baena
- Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06070 USA
| | - Shujun Ge
- Blood-Brain Barrier Laboratory, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06070 USA
- Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06070 USA
| | - Xi Jiang
- Blood-Brain Barrier Laboratory, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06070 USA
- Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06070 USA
| | - Evan R. Jellison
- Department of Immunology, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06070 USA
| | - Timothy Kiprono
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06070 USA
| | - Dritan Agalliu
- Department of Pathology and Cell Biology, Columbia University School of Medicine, 630 W 168th St, New York, NY 10032 USA
| | - Joel S. Pachter
- Blood-Brain Barrier Laboratory, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06070 USA
- Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06070 USA
| |
Collapse
|
67
|
Schmidt EP, Kuebler WM, Lee WL, Downey GP. Adhesion Molecules: Master Controllers of the Circulatory System. Compr Physiol 2016; 6:945-73. [PMID: 27065171 DOI: 10.1002/cphy.c150020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This manuscript will review our current understanding of cellular adhesion molecules (CAMs) relevant to the circulatory system, their physiological role in control of vascular homeostasis, innate and adaptive immune responses, and their importance in pathophysiological (disease) processes such as acute lung injury, atherosclerosis, and pulmonary hypertension. This is a complex and rapidly changing area of research that is incompletely understood. By design, we will begin with a brief overview of the structure and classification of the major groups of adhesion molecules and their physiological functions including cellular adhesion and signaling. The role of specific CAMs in the process of platelet aggregation and hemostasis and leukocyte adhesion and transendothelial migration will be reviewed as examples of the complex and cooperative interplay between CAMs during physiological and pathophysiological processes. The role of the endothelial glycocalyx and the glycobiology of this complex system related to inflammatory states such as sepsis will be reviewed. We will then focus on the role of adhesion molecules in the pathogenesis of specific disease processes involving the lungs and cardiovascular system. The potential of targeting adhesion molecules in the treatment of immune and inflammatory diseases will be highlighted in the relevant sections throughout the manuscript.
Collapse
Affiliation(s)
- Eric P Schmidt
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Wolfgang M Kuebler
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Departments of Surgery and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Warren L Lee
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Division of Respirology and the Interdepartmental Division of Critical Care Medicine, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gregory P Downey
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Departments of Medicine, Pediatrics, and Biomedical Research, National Jewish Health, Denver, Colorado, USA
- Departments of Medicine, and Immunology and Microbiology, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
68
|
Youngren-Ortiz SR, Gandhi NS, España-Serrano L, Chougule MB. Aerosol Delivery of siRNA to the Lungs. Part 1: Rationale for Gene Delivery Systems. KONA : POWDER SCIENCE AND TECHNOLOGY IN JAPAN 2016; 33:63-85. [PMID: 27081214 PMCID: PMC4829385 DOI: 10.14356/kona.2016014] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
This article reviews the pulmonary route of administration, aerosol delivery devices, characterization of pulmonary drug delivery systems, and discusses the rationale for inhaled delivery of siRNA. Diseases with known protein malfunctions may be mitigated through the use of siRNA therapeutics. The inhalation route of administration provides local delivery of siRNA therapeutics for the treatment of various pulmonary diseases, however barriers to pulmonary delivery and intracellular delivery of siRNA exists. siRNA loaded nanocarriers can be used to overcome the barriers associated with the pulmonary route, such as anatomical barriers, mucociliary clearance, and alveolar macrophage clearance. Apart from naked siRNA aerosol delivery, previously studied siRNA carrier systems comprise of lipidic, polymeric, peptide, or inorganic origin. Such siRNA delivery systems formulated as aerosols can be successfully delivered via an inhaler or nebulizer to the pulmonary region. Preclinical animal investigations of inhaled siRNA therapeutics rely on intratracheal and intranasal siRNA and siRNA nanocarrier delivery. Aerosolized siRNA delivery systems may be characterized using in vitro techniques, such as dissolution test, inertial cascade impaction, delivered dose uniformity assay, laser diffraction, and laser Doppler velocimetry. The ex vivo techniques used to characterize pulmonary administered formulations include the isolated perfused lung model. In vivo techniques like gamma scintigraphy, 3D SPECT, PET, MRI, fluorescence imaging and pharmacokinetic/pharmacodynamics analysis may be used for evaluation of aerosolized siRNA delivery systems. The use of inhalable siRNA delivery systems encounters barriers to their delivery, however overcoming the barriers while formulating a safe and effective delivery system will offer unique advances to the field of inhaled medicine.
Collapse
Affiliation(s)
- Susanne R. Youngren-Ortiz
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, 200 West Kawili Street, Hilo, Hawaii 96720, USA
| | - Nishant S. Gandhi
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, 200 West Kawili Street, Hilo, Hawaii 96720, USA
| | - Laura España-Serrano
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, 200 West Kawili Street, Hilo, Hawaii 96720, USA
| | - Mahavir B. Chougule
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, 200 West Kawili Street, Hilo, Hawaii 96720, USA
- Natural Products and Experimental Therapeutics Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii 96813, USA
| |
Collapse
|
69
|
Petrovich E, Feigelson SW, Stoler-Barak L, Hatzav M, Solomon A, Bar-Shai A, Ilan N, Li JP, Engelhardt B, Vlodavsky I, Alon R. Lung ICAM-1 and ICAM-2 support spontaneous intravascular effector lymphocyte entrapment but are not required for neutrophil entrapment or emigration inside endotoxin-inflamed lungs. FASEB J 2016; 30:1767-78. [PMID: 26823454 DOI: 10.1096/fj.201500046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 12/22/2015] [Indexed: 11/11/2022]
Abstract
The pulmonary vasculature constitutively expresses the integrin lymphocyte function-associated antigen-1 ligands intercellular adhesion molecule (ICAM)-1 and -2. In this study, effector T cells were temporarily entrapped by the lung vasculature on their way to inflamed lymph nodes, and this entrapment was strongly reduced in ICAM-1 and -2 double-deficient mice (79 and 86% reduction for CD8(+) and CD4(+) effectors, respectively, compared with wild-type mice). Although the pulmonary vasculature has been suggested to be masked by the heparan sulfate-containing glycocalyx, which is susceptible to heparanase-mediated shedding, lung and lymphocyte heparanase have been found to be unnecessary for this entrapment. Systemic LPS induced rapid neutrophil entrapment in the lung vasculature, but in contrast to T-cell entrapment, this sequestration was ICAM-1, ICAM-2, and heparanase independent. Furthermore, neutrophil migration into the bronchoalveolar space induced by LPS inhalation and LPS-induced leakage of red blood cells into this space were not dependent on lung ICAMs or heparanase activity. Nevertheless, heparanase was critical for neutrophil accumulation in smoke-exposed lungs. Our results indicate that, whereas T cells use ICAM-1 and -2 for temporary pulmonary entrapment, neutrophils get sequestered and extravasate into inflamed lungs independent of ICAMs. This is the first demonstration that the pulmonary vasculature is differentially recognized by T cells and neutrophils.-Petrovich, E., Feigelson, S. W., Stoler-Barak, L., Hatzav, M., Solomon, A., Bar-Shai, A., Ilan, N., Li, J.-P., Engelhardt, B., Vlodavsky, I., Alon, R. Lung ICAM-1 and ICAM-2 support spontaneous intravascular effector lymphocyte entrapment but are not required for neutrophil entrapment or emigration inside endotoxin-inflamed lungs.
Collapse
Affiliation(s)
- Ekaterina Petrovich
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Sara W Feigelson
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Liat Stoler-Barak
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Miki Hatzav
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Adam Solomon
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Amir Bar-Shai
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Neta Ilan
- Cancer and Vascular Biology Research Center, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, University of Uppsala, Uppsala, Sweden; and
| | | | - Israel Vlodavsky
- Cancer and Vascular Biology Research Center, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ronen Alon
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel;
| |
Collapse
|
70
|
Abstract
Extracellular matrix (ECM) is a tissue-specific macromolecular structure that provides physical support to tissues and is essential for normal organ function. In the lung, ECM plays an active role in shaping cell behavior both in health and disease by virtue of the contextual clues it imparts to cells. Qualities including dimensionality, molecular composition, and intrinsic stiffness all promote normal function of the lung ECM. Alterations in composition and/or modulation of stiffness of the focally injured or diseased lung ECM microenvironment plays a part in reparative processes performed by fibroblasts. Under conditions of remodeling or in disease states, inhomogeneous stiffening (or softening) of the pathologic ECM may both precede modifications in cell behavior and be a result of disease progression. The ability of ECM to stimulate further ECM production by fibroblasts and drive disease progression has potentially significant implications for mesenchymal stromal cell-based therapies; in the setting of pathologic ECM stiffness or composition, the therapeutic intent of progenitor cells may be subverted. Taken together, current data suggest that lung ECM actively contributes to health and disease; thus, mediators of cell-ECM signaling or factors that influence ECM stiffness may represent viable therapeutic targets in many lung disorders.
Collapse
|
71
|
Abstract
Submucosal glands contribute to airway surface liquid (ASL), a film that protects all airway surfaces. Glandular mucus comprises electrolytes, water, the gel-forming mucin MUC5B, and hundreds of different proteins with diverse protective functions. Gland volume per unit area of mucosal surface correlates positively with impaction rate of inhaled particles. In human main bronchi, the volume of the glands is ∼ 50 times that of surface goblet cells, but the glands diminish in size and frequency distally. ASL and its trapped particles are removed from the airways by mucociliary transport. Airway glands have a tubuloacinar structure, with a single terminal duct, a nonciliated collecting duct, then branching secretory tubules lined with mucous cells and ending in serous acini. They allow for a massive increase in numbers of mucus-producing cells without replacing surface ciliated cells. Active secretion of Cl(-) and HCO3 (-) by serous cells produces most of the fluid of gland secretions. Glands are densely innervated by tonically active, mutually excitatory airway intrinsic neurons. Most gland mucus is secreted constitutively in vivo, with large, transient increases produced by emergency reflex drive from the vagus. Elevations of [cAMP]i and [Ca(2+)]i coordinate electrolyte and macromolecular secretion and probably occur together for baseline activity in vivo, with cholinergic elevation of [Ca(2+)]i being mainly responsive for transient increases in secretion. Altered submucosal gland function contributes to the pathology of all obstructive diseases, but is an early stage of pathogenesis only in cystic fibrosis.
Collapse
Affiliation(s)
- Jonathan H Widdicombe
- Department of Physiology and Membrane Biology, University of California-Davis, Davis, California; and Department of Psychology and Cystic Fibrosis Research Laboratory, Stanford University, Stanford, California
| | - Jeffrey J Wine
- Department of Physiology and Membrane Biology, University of California-Davis, Davis, California; and Department of Psychology and Cystic Fibrosis Research Laboratory, Stanford University, Stanford, California
| |
Collapse
|
72
|
Le NPK, Channabasappa S, Hossain M, Liu L, Singh B. Leukocyte-specific protein 1 regulates neutrophil recruitment in acute lung inflammation. Am J Physiol Lung Cell Mol Physiol 2015; 309:L995-1008. [PMID: 26320151 DOI: 10.1152/ajplung.00068.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 08/25/2015] [Indexed: 01/21/2023] Open
Abstract
The mechanisms of excessive migration of activated neutrophils into inflamed lungs, credited with tissue damage, are not fully understood. We explored the hitherto unknown expression of leukocyte-specific protein 1 (LSP1) in human and mouse lungs and neutrophils and examined its role in neutrophil migration in acute lung inflammation. Autopsied septic human lungs showed increased LSP1 labeling in epithelium, endothelium, and leukocytes, including in their nuclei compared with normal lungs. We induced acute lung inflammation through intranasal administration of E. coli lipopolysaccharide (LPS) (80 μg) in LSP1-deficient (Lsp1(-/-)) and wild-type (WT) 129/SvJ mice. Immunocytochemistry and Western blots showed increased expression of LSP1 and phosphorylated LSP1 in lungs of LPS-treated WT mice. Histology showed more congestion, inflammation, and Gr-1(+) neutrophils in lung of WT mice than Lsp1(-/-) mice. LPS-treated WT mice had significantly more neutrophils in bronchoalveolar lavage (BAL) and myeloperoxidase levels in lungs compared with Lsp1(-/-) mice. However, there were no differences in lung tissue and BAL concentrations of keratinocyte-derived chemokine, monocyte chemoattractant protein-1, macrophage inflammatory protein-1α and -1β, vascular permeability, and phosphorylated p38 MAPK between LPS-treated WT and Lsp1(-/-) mice, whereas TNF-α concentration was higher in BAL fluid from LPS-treated WT. Immunoelectron microscopy showed increased LSP1 in the nuclei of LPS-treated neutrophils. We also found increased levels of phosphorylated LSP1 associated with plasma membrane, nucleus, and cytosol at various times after LPS treatment of murine bone marrow-derived neutrophils, suggesting its role in modulation of neutrophil cytoskeleton and the membrane. These data collectively show increased expression of LSP1 in inflamed mouse and human lungs and its role in neutrophil recruitment and lung inflammation.
Collapse
Affiliation(s)
- Nguyen Phuong Khanh Le
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Ho Chi Minh City, Vietnam
| | - Shankaramurthy Channabasappa
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Mokarram Hossain
- Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; and
| | - Lixin Liu
- Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; and
| | - Baljit Singh
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada;
| |
Collapse
|
73
|
Ekpenyong AE, Toepfner N, Chilvers ER, Guck J. Mechanotransduction in neutrophil activation and deactivation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015. [PMID: 26211453 DOI: 10.1016/j.bbamcr.2015.07.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Mechanotransduction refers to the processes through which cells sense mechanical stimuli by converting them to biochemical signals and, thus, eliciting specific cellular responses. Cells sense mechanical stimuli from their 3D environment, including the extracellular matrix, neighboring cells and other mechanical forces. Incidentally, the emerging concept of mechanical homeostasis,long term or chronic regulation of mechanical properties, seems to apply to neutrophils in a peculiar manner, owing to neutrophils' ability to dynamically switch between the activated/primed and deactivated/deprimed states. While neutrophil activation has been known for over a century, its deactivation is a relatively recent discovery. Even more intriguing is the reversibility of neutrophil activation and deactivation. We review and critically evaluate recent findings that suggest physiological roles for neutrophil activation and deactivation and discuss possible mechanisms by which mechanical stimuli can drive the oscillation of neutrophils between the activated and resting states. We highlight several molecules that have been identified in neutrophil mechanotransduction, including cell adhesion and transmembrane receptors, cytoskeletal and ion channel molecules. The physiological and pathophysiological implications of such mechanically induced signal transduction in neutrophils are highlighted as a basis for future work. This article is part of a Special Issue entitled: Mechanobiology.
Collapse
Affiliation(s)
- Andrew E Ekpenyong
- Department of Physics, Creighton University, Omaha, NE 68178, USA; Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Nicole Toepfner
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany; Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Edwin R Chilvers
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Papworth Hospitals, Cambridge CB2 0QQ, UK
| | - Jochen Guck
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
74
|
Hallmann R, Zhang X, Di Russo J, Li L, Song J, Hannocks MJ, Sorokin L. The regulation of immune cell trafficking by the extracellular matrix. Curr Opin Cell Biol 2015; 36:54-61. [PMID: 26189064 DOI: 10.1016/j.ceb.2015.06.006] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/04/2015] [Accepted: 06/27/2015] [Indexed: 12/11/2022]
Abstract
The extracellular matrix (ECM) comes in different structural forms and biochemical compositions, which determine both its biophysical properties and its ability to convey specific signals to immune cells encountering or navigating through it. Traditionally, the role of the individual ECM molecules on cell migration has been investigated independent of considerations such as the tension/mechanical strength constituted by the ECM. However, more recently, this aspect has attracted considerable attention and data suggest that rigidity and molecular signals derived from the ECM define the mode of cell migration. We here review the different types of ECM encountered by migrating immune cells in vivo, as well as current information on how both molecular components of the ECM and their supramolecular structure can impact on modes of immune cell migration.
Collapse
Affiliation(s)
- Rupert Hallmann
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Muenster, Germany
| | - Xueli Zhang
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Muenster, Germany
| | - Jacopo Di Russo
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Muenster, Germany
| | - Lixia Li
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Muenster, Germany
| | - Jian Song
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Muenster, Germany
| | - Melanie-Jane Hannocks
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Muenster, Germany
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Muenster, Germany.
| |
Collapse
|
75
|
Sharma P, Bansal A, Sharma PC. RNA-seq-based transcriptome profiling reveals differential gene expression in the lungs of Sprague-Dawley rats during early-phase acute hypobaric hypoxia. Mol Genet Genomics 2015; 290:2225-40. [PMID: 26050109 DOI: 10.1007/s00438-015-1064-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 05/02/2015] [Indexed: 12/19/2022]
Abstract
Individuals subjected to hypobaric hypoxia at high altitudes may exhibit differential physiological responses in terms of susceptibility and tolerance to the development of hypoxia-related disorders. We studied early-phase gene expression in the lungs of Sprague-Dawley rats exhibiting such differential physiological responses after exposure to acute hypobaric hypoxia for 1 h at a simulated altitude of 9144 m. RNA-seq transcriptome profiling of lung tissues revealed differential gene expression in tolerant and susceptible groups, subsequently validated by qRT-PCR for ten selected differentially expressed genes. The gene expression pattern indicated hypometabolism and negative regulation of vasoconstriction in all groups except susceptible rats, coupled with altered MAPK, p53 and JAK-STAT signaling. Upregulation of early-phase response genes including Dusp1 (dual specificity phosphatase), Cdkn1a (cyclin-dependent kinase inhibitor 1A), Txnip (thioredoxin-interacting protein), Rgs1 (regulator of G-protein signaling 1) and Rgs2 (regulator of G-protein signaling 2) in susceptible rats indicated a progression toward growth arrest and apoptosis. Enhanced expression of cell adhesion molecules, wound healing and repair bioprocesses was observed in tolerant males. Upregulated Kcnj15 (potassium inwardly rectifying channel subfamily j membrane 15) and Vsig4 (V-set and Ig domain containing 4) variants in tolerant females suggested adaptation to hypoxia possibly by fluid reabsorption to avoid edematous conditions and suppression of T cell proliferation to avoid acute lung inflammation. Our study might help in understanding the molecular-physiological mechanisms associated with progressive damage in the lung tissues of susceptible and tissue-protective measures in tolerant rats during acute hypobaric hypoxia.
Collapse
Affiliation(s)
- Priyanka Sharma
- University School of Biotechnology (USBT), Guru Gobind Singh Indraprastha University, Sector 16C, Dwarka, New Delhi, 110078, India
| | - Anju Bansal
- Experimental Biology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organisation (DRDO), Timarpur, Lucknow Road, New Delhi, India
| | - Prakash Chand Sharma
- University School of Biotechnology (USBT), Guru Gobind Singh Indraprastha University, Sector 16C, Dwarka, New Delhi, 110078, India.
| |
Collapse
|
76
|
Bai X, Fan L, He T, Jia W, Yang L, Zhang J, Liu Y, Shi J, Su L, Hu D. SIRT1 protects rat lung tissue against severe burn-induced remote ALI by attenuating the apoptosis of PMVECs via p38 MAPK signaling. Sci Rep 2015; 5:10277. [PMID: 25992481 PMCID: PMC4445725 DOI: 10.1038/srep10277] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/08/2015] [Indexed: 12/14/2022] Open
Abstract
Silent information regulator type-1 (SIRT1) has been reported to be involved in the
cardiopulmonary protection. However, its role in the pathogenesis of burn-induced
remote acute lung injury (ALI) is currently unknown. The present study aims to
investigate the role of SIRT1 in burn-induced remote ALI and the involved signaling
pathway. We observed that SIRT1 expression in rat lung tissue after burn injury
appeared an increasing trend after a short period of suppression. The upregulation
of SIRT1 stimulated by resveratrol exhibited remission of histopathologic changes,
reduction of cell apoptosis, and downregulation of pro-inflammatory cytokines in rat
pulmonary tissues suffering from severe burn. We next used primary pulmonary
microvascular endothelial cells (PMVECs) challenged by burn serum (BS) to simulate
in vivo rat lung tissue after burn injury, and found that BS
significantly suppressed SIRT1 expression, increased cell apoptosis, and activated
p38 MAPK signaling. The use of resveratrol reversed these effects, while knockdown
of SIRT1 by shRNA further augmented BS-induced increase of cell apoptosis and
activation of p38 MAPK. Taken together, these results indicate that SIRT1 might
protect lung tissue against burn-induced remote ALI by attenuating PMVEC apoptosis
via p38 MAPK signaling, suggesting its potential therapeutic effects on the
treatment of ALI.
Collapse
Affiliation(s)
- Xiaozhi Bai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lei Fan
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ting He
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wenbin Jia
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Longlong Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jun Zhang
- Department of Burn and Plastic Surgery, No.205 Hospital of Chinese People's Liberation Army, Jinzhou, Liaoning, China
| | - Yang Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jihong Shi
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Linlin Su
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
77
|
Yonker LM, Cigana C, Hurley BP, Bragonzi A. Host-pathogen interplay in the respiratory environment of cystic fibrosis. J Cyst Fibros 2015; 14:431-439. [PMID: 25800687 DOI: 10.1016/j.jcf.2015.02.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 02/11/2015] [Accepted: 02/19/2015] [Indexed: 01/01/2023]
Abstract
Significant advances have been made in the understanding of disease progression in cystic fibrosis (CF), revealing a complex interplay between host and pathogenic organisms. The diverse CF microbiota within the airway activates an aberrant immune response that is ineffective in clearing infection. An appreciation of how the CF host immune system interacts with these organisms is crucial to understanding the pathogenesis of CF pulmonary disease. Here we discuss the microbial complexity present in the lungs of individuals with CF, review emerging concepts of innate and adaptive immune responses to pathogens that chronically inhabit the CF lung, and discuss therapies that target the aberrant inflammatory response that characterizes CF. A greater understanding of the underlying mechanisms will shed light on pathogenesis and guide more targeted therapies in the future that serve to reduce infection, minimize lung pathology, and improve the quality of life for patients with CF.
Collapse
Affiliation(s)
- Lael M Yonker
- Mucosal Immunology & Biology Research Center, Pediatrics, Harvard Medical School, Massachusetts General Hospital for Children , Charlestown, MA, U.S.A
| | - Cristina Cigana
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Bryan P Hurley
- Mucosal Immunology & Biology Research Center, Pediatrics, Harvard Medical School, Massachusetts General Hospital for Children , Charlestown, MA, U.S.A
| | - Alessandra Bragonzi
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
78
|
Ambroggio L, Test M, Metlay JP, Graf TR, Blosky MA, Macaluso M, Shah SS. Adjunct Systemic Corticosteroid Therapy in Children With Community-Acquired Pneumonia in the Outpatient Setting. J Pediatric Infect Dis Soc 2015; 4:21-7. [PMID: 26407353 PMCID: PMC5965878 DOI: 10.1093/jpids/piu017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 02/03/2014] [Indexed: 11/14/2022]
Abstract
BACKGROUND The role of adjunct systemic corticosteroid therapy in children with community-acquired pneumonia (CAP) is not known. The objective was to determine the association between adjunct systemic corticosteroid therapy and treatment failure in children who received antibiotics for treatment of CAP in the outpatient setting. METHODS The study included a retrospective cohort study of children, aged 1-18 years, with a diagnosis of CAP who were managed at an outpatient practice affiliated with Geisinger Health System from January 1, 2008 to January 31, 2010. The primary exposure was the receipt of adjunct corticosteroid therapy. The primary outcome was treatment failure defined as a respiratory-associated follow-up within 14 days of diagnosis in which the participant received a change in antibiotic therapy. The probability of receiving adjunct systemic corticosteroid therapy was calculated using a matched propensity score. A multivariable conditional logistic regression model was used to estimate the association between adjunct corticosteroids and treatment failure. RESULTS Of 2244 children with CAP, 293 (13%) received adjunct corticosteroids, 517 (23%) had underlying asthma, and 624 (28%) presented with wheezing. Most patients received macrolide monotherapy for their CAP diagnosis (n = 1329; 59%). Overall, treatment failure was not associated with adjunct corticosteroid treatment (odds ratio [OR], 1.72; 95% confidence interval [CI], 0.93 and 3.19), but the association was statistically significant among patients with no history of asthma (OR, 2.38; 95% CI, 1.03 and 5.52), with no statistical association among patients with a history of asthma. CONCLUSION Adjunct corticosteroid therapy was associated with treatment failure among children diagnosed with CAP who did not have underlying asthma.
Collapse
Affiliation(s)
- Lilliam Ambroggio
- Division of Hospital Medicine,Division of Biostatistics and Epidemiology,Department of Pediatrics, the University of Cincinnati College of Medicine, Ohio
| | | | - Joshua P. Metlay
- General Medicine Division, Massachusetts General Hospital, Boston
| | - Thomas R. Graf
- Population Health, Geisinger Health System, Danville, Pennsylvania
| | - Mary Ann Blosky
- Population Health, Geisinger Health System, Danville, Pennsylvania
| | - Maurizio Macaluso
- Division of Biostatistics and Epidemiology,Department of Pediatrics, the University of Cincinnati College of Medicine, Ohio
| | - Samir S. Shah
- Division of Hospital Medicine,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center,Department of Pediatrics, the University of Cincinnati College of Medicine, Ohio
| |
Collapse
|
79
|
Cheng Y, Zhang Y, Song H, Feng J. Intercellular adhesion molecule-1 expression in the hippocampal CA1 region of hyperlipidemic rats with chronic cerebral ischemia. Neural Regen Res 2015; 7:1312-7. [PMID: 25657661 PMCID: PMC4308801 DOI: 10.3969/j.issn.1673-5374.2012.17.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/21/2012] [Indexed: 11/18/2022] Open
Abstract
Chronic cerebral ischemia is a pathological process in many cerebrovascular diseases and it is induced by long-term hyperlipidemia, hypertension and diabetes mellitus. After being fed a high-fat diet for 4 weeks, rats were subjected to permanent occlusion of bilateral common carotid arteries to establish rat models of chronic cerebral ischemia with hyperlipidemia. Intercellular adhesion molecule-1 expression in rat hippocampal CA1 region was determined to better understand the mechanism underlying the effects of hyperlipidemia on chronic cerebral ischemia. Water maze test results showed that the cognitive function of rats with hyperlipidemia or chronic cerebral ischemia, particularly in rats with hyperlipidemia combined with chronic cerebral ischemia, gradually decreased between 1 and 4 months after occlusion of the bilateral common carotid arteries. This correlated with pathological changes in the hippocampal CA1 region as detected by hematoxylin-eosin staining. Immunohistochemical staining showed that intercellular adhesion molecule-1 expression in the hippocampal CA1 region was noticeably increased in rats with hyperlipidemia or chronic cerebral ischemia, in particular in rats with hyperlipidemia combined with chronic cerebral ischemia. These findings suggest that hyperlipidemia aggravates chronic cerebral ischemia-induced neurological damage and cognitive impairment in the rat hippocampal CA1 region, which may be mediated, at least in part, by up-regulated expression of intercellular adhesion molecule-1.
Collapse
Affiliation(s)
- Yingying Cheng
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Ying Zhang
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Hongmei Song
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Jiachun Feng
- Department of Neurology, The First Bethune Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
80
|
Luyts K, Napierska D, Dinsdale D, Klein SG, Serchi T, Hoet PH. A coculture model of the lung–blood barrier: The role of activated phagocytic cells. Toxicol In Vitro 2015; 29:234-41. [DOI: 10.1016/j.tiv.2014.10.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 10/17/2014] [Accepted: 10/28/2014] [Indexed: 01/16/2023]
|
81
|
Mao P, Wu S, Li J, Fu W, He W, Liu X, Slutsky AS, Zhang H, Li Y. Human alveolar epithelial type II cells in primary culture. Physiol Rep 2015; 3:e12288. [PMID: 25677546 PMCID: PMC4393197 DOI: 10.14814/phy2.12288] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 12/21/2014] [Accepted: 01/08/2015] [Indexed: 01/13/2023] Open
Abstract
Alveolar epithelial type II (AEII) cells are a key structure and defender in the lung but also are the targets in many lung diseases, including acute respiratory distress syndrome, ventilator-induced lung injury, and pulmonary fibrosis. We sought to establish an optimized method for high yielding and long maintenance of characteristics of primary human AEII cells to facilitate the investigation of the mechanisms of lung diseases at the cellular and molecular levels. Adult human peripheral normal lung tissues of oncologic patients undergoing lung resection were collected. The AEII cells were isolated and identified by the expression of pro-surfactant protein (SP)C, epithelial sodium channel (αENaC) and cytokeratin (CK)-8, the lamellar bodies specific for AEII cells, and confirmed by the histology using electron microscopy. The phenotype of AEII cells was characterized by the expression of surfactant proteins (SP-A, SP-B, SP-C, SP-D), CK-8, KL-6, αENaC, and aquaporin (AQP)-3, which was maintained over 20 days. The biological activity of the primary human AEII cells producing SP-C, cytokines, and intercellular adhesion molecule-1 was vigorous in response to stimulation with tumor necrosis factor-α. We have modified previous methods and optimized a method for isolation of high purity and long maintenance of the human AEII cell phenotype in primary culture. This method provides an important tool for studies aiming at elucidating the molecular mechanisms of lung diseases exclusively in AEII cells.
Collapse
Affiliation(s)
- Pu Mao
- State Key Laboratory of Respiratory Diseases and Guangzhou Institute of Respiratory DiseasesGuangzhou, Guangdong, China
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Guangdong, China
| | - Songling Wu
- State Key Laboratory of Respiratory Diseases and Guangzhou Institute of Respiratory DiseasesGuangzhou, Guangdong, China
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Guangdong, China
| | - Jianchun Li
- State Key Laboratory of Respiratory Diseases and Guangzhou Institute of Respiratory DiseasesGuangzhou, Guangdong, China
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Guangdong, China
| | - Wei Fu
- State Key Laboratory of Respiratory Diseases and Guangzhou Institute of Respiratory DiseasesGuangzhou, Guangdong, China
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Guangdong, China
| | - Weiqun He
- State Key Laboratory of Respiratory Diseases and Guangzhou Institute of Respiratory DiseasesGuangzhou, Guangdong, China
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Guangdong, China
| | - Xiaoqing Liu
- State Key Laboratory of Respiratory Diseases and Guangzhou Institute of Respiratory DiseasesGuangzhou, Guangdong, China
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Guangdong, China
| | - Arthur S Slutsky
- State Key Laboratory of Respiratory Diseases and Guangzhou Institute of Respiratory DiseasesGuangzhou, Guangdong, China
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Guangdong, China
- Keenan Research Centre for Biomedical Science of St. Michael's HospitalToronto, Ontario, Canada
- Department of Medicine, University of TorontoToronto, Ontario, Canada
| | - Haibo Zhang
- State Key Laboratory of Respiratory Diseases and Guangzhou Institute of Respiratory DiseasesGuangzhou, Guangdong, China
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Guangdong, China
- Keenan Research Centre for Biomedical Science of St. Michael's HospitalToronto, Ontario, Canada
- Department of Medicine, University of TorontoToronto, Ontario, Canada
- Department of Anesthesia, University of TorontoToronto, Ontario, Canada
- Department of Physiology, University of TorontoToronto, Ontario, Canada
| | - Yimin Li
- State Key Laboratory of Respiratory Diseases and Guangzhou Institute of Respiratory DiseasesGuangzhou, Guangdong, China
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, Guangdong, China
| |
Collapse
|
82
|
Bogdani M, Korpos E, Simeonovic CJ, Parish CR, Sorokin L, Wight TN. Extracellular matrix components in the pathogenesis of type 1 diabetes. Curr Diab Rep 2014; 14:552. [PMID: 25344787 PMCID: PMC4238291 DOI: 10.1007/s11892-014-0552-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Type 1 diabetes (T1D) results from progressive immune cell-mediated destruction of pancreatic β cells. As immune cells migrate into the islets, they pass through the extracellular matrix (ECM). This ECM is composed of different macromolecules localized to different compartments within and surrounding islets; however, the involvement of this ECM in the development of human T1D is not well understood. Here, we summarize our recent findings from human and mouse studies illustrating how specific components of the islet ECM that constitute basement membranes and interstitial matrix of the islets, and surprisingly, the intracellular composition of islet β cells themselves, are significantly altered during the pathogenesis of T1D. Our focus is on the ECM molecules laminins, collagens, heparan sulfate/heparan sulfate proteoglycans, and hyaluronan, as well as on the enzymes that degrade these ECM components. We propose that islet and lymphoid tissue ECM composition and organization are critical to promoting immune cell activation, islet invasion, and destruction of islet β cells in T1D.
Collapse
Affiliation(s)
- Marika Bogdani
- Matrix Biology Program, Benaroya Research Institute, 1201 Ninth Avenue, Seattle, WA 98101 Ph: 206-287-5666, Fax: 206-342-6567
| | - Eva Korpos
- Institute of Physiological Chemistry and Pathobiochemistry,Cells-in-Motion Cluster of Excellence (EXC 1003 – CiM), University of Münster, Münster, Germany
| | - Charmaine J. Simeonovic
- Diabetes/Transplantation Immunobiology Laboratory, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601 Australia
| | - Christopher R. Parish
- Cancer and Vascular Biology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry,Cells-in-Motion Cluster of Excellence (EXC 1003 – CiM), University of Münster, Münster, Germany
| | - Thomas N. Wight
- Matrix Biology Program, Benaroya Research Institute, 1201 Ninth Avenue, Seattle, WA 98101 Ph: 206-287-5666, Fax: 206-342-6567
- Corresponding Author: Thomas N. Wight, PhD
| |
Collapse
|
83
|
Orme IM. Vaccines to prevent tuberculosis infection rather than disease: Physiological and immunological aspects. Tuberculosis (Edinb) 2014; 101:210-216. [PMID: 25500316 DOI: 10.1016/j.tube.2014.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 09/02/2014] [Accepted: 10/22/2014] [Indexed: 12/11/2022]
Abstract
There is increasing enthusiasm and optimism that a vaccine could be developed that prevents infection rather than disease. In this article I discuss the fact that despite this optimism nothing has been produced so far that seems to have this capability, and moreover even the borderline between when infection ends and disease begins has not even been defined. To be effective such a vaccine, or at least the immunity it would generate, would have to work within the confines of the pulmonary physiological systems, which are complex. To date much of the emphasis here has turned away from T cell mediated immunity and towards establishing specific antibodies in the lungs. Here, I argue that with the exception of a possible exclusionary function, most claims of a protective role of antibody are completely over-blown. Finally, even if we had a potential "anti-infection" vaccine, how would we test and validate it?
Collapse
Affiliation(s)
- Ian M Orme
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
84
|
Hebbel RP. Ischemia-reperfusion injury in sickle cell anemia: relationship to acute chest syndrome, endothelial dysfunction, arterial vasculopathy, and inflammatory pain. Hematol Oncol Clin North Am 2014; 28:181-98. [PMID: 24589261 DOI: 10.1016/j.hoc.2013.11.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Ischemia-reperfusion (I/R) physiology, also called reperfusion injury, instigates vascular and tissue injury in human disease states. This review describes why sickle cell anemia should be conceptualized in this fashion and how I/R physiology explains the genesis of characteristic aspects of vascular pathobiology and clinical disease in sickle cell anemia. The nature of I/R and its relevance to sickle cell anemia are discussed, with an emphasis on the acute chest syndrome, endothelial dysfunction with aberrant vasoregulation, circle of Willis vasculopathy, and inflammatory pain. Viewing sickle disease from this perspective elucidates defining pathophysiology and identifies a host of novel potential therapeutic targets.
Collapse
Affiliation(s)
- Robert P Hebbel
- Division of Hematology-Oncology-Transplantation, Department of Medicine, University of Minnesota Medical School, 420 Delaware Street South East, Mayo Mail Code 480, Minneapolis, MN 55455, USA.
| |
Collapse
|
85
|
Ortiz-Muñoz G, Mallavia B, Bins A, Headley M, Krummel MF, Looney MR. Aspirin-triggered 15-epi-lipoxin A4 regulates neutrophil-platelet aggregation and attenuates acute lung injury in mice. Blood 2014; 124:2625-34. [PMID: 25143486 PMCID: PMC4208278 DOI: 10.1182/blood-2014-03-562876] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 08/05/2014] [Indexed: 01/14/2023] Open
Abstract
Evidence is emerging that platelets are major contributors to innate immune responses in conditions such as acute lung injury (ALI). Platelets form heterotypic aggregates with neutrophils, and we hypothesized that lipoxin mediators regulate formation of neutrophil-platelet aggregates (NPA) and that NPA significantly contribute to ALI. Lipopolysaccharide (LPS)-induced lung injury was accompanied by platelet sequestration, activation, intra-alveolar accumulation, and NPA formation within both blood and alveolar compartments. Using lung intravital microscopy, we observed the dynamic formation of NPA during physiologic conditions, which sharply increased with ALI. Aspirin (ASA) treatment significantly reduced lung platelet sequestration and activation, NPA formation, and lung injury. ASA treatment increased levels of ASA-triggered lipoxin (ATL; 15-epi-lipoxin A4), and blocking the lipoxin A4 receptor (ALX) with a peptide antagonist (Boc2) or using ALX knockouts (Fpr2/3(-/-)) reversed this protection. LPS increased NPA formation in vitro, which was reduced by ATL, and engagement of ALX by ATL on both neutrophils and platelets was necessary to prevent aggregation. In a model of transfusion-related acute lung injury (TRALI), Boc2 also reversed ASA protection, and treatment with ATL in both LPS and TRALI models protected from ALI. We conclude that ATL regulates neutrophil-platelet aggregation and that platelet-neutrophil interactions are a therapeutic target in lung injury.
Collapse
Affiliation(s)
| | - Beñat Mallavia
- Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Adriaan Bins
- Department of Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | - Mark R Looney
- Department of Medicine, University of California, San Francisco, San Francisco, CA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
86
|
Xu F, Diao R, Liu J, Kang Y, Wang X, Shi L. Curcumin attenuatesstaphylococcus aureus-induced acute lung injury. CLINICAL RESPIRATORY JOURNAL 2014; 9:87-97. [PMID: 24460792 DOI: 10.1111/crj.12113] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 01/13/2014] [Accepted: 01/21/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Feng Xu
- Department of Infectious Diseases; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou China
- Department of Respiratory Medicine; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou China
| | - Ran Diao
- Department of Respiratory Medicine; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou China
- Center for Allergy; Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou China
| | - Jin Liu
- Department of Infectious Diseases; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou China
| | - Yanhua Kang
- Division of Immunology; Hangzhou Normal University; Hangzhou China
| | - Xuanding Wang
- Department of Respiratory Medicine; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou China
| | - Liyun Shi
- Division of Immunology; Hangzhou Normal University; Hangzhou China
| |
Collapse
|
87
|
Kusek ME, Pazos MA, Pirzai W, Hurley BP. In vitro coculture assay to assess pathogen induced neutrophil trans-epithelial migration. J Vis Exp 2014:e50823. [PMID: 24430378 DOI: 10.3791/50823] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Mucosal surfaces serve as protective barriers against pathogenic organisms. Innate immune responses are activated upon sensing pathogen leading to the infiltration of tissues with migrating inflammatory cells, primarily neutrophils. This process has the potential to be destructive to tissues if excessive or held in an unresolved state. Cocultured in vitro models can be utilized to study the unique molecular mechanisms involved in pathogen induced neutrophil trans-epithelial migration. This type of model provides versatility in experimental design with opportunity for controlled manipulation of the pathogen, epithelial barrier, or neutrophil. Pathogenic infection of the apical surface of polarized epithelial monolayers grown on permeable transwell filters instigates physiologically relevant basolateral to apical trans-epithelial migration of neutrophils applied to the basolateral surface. The in vitro model described herein demonstrates the multiple steps necessary for demonstrating neutrophil migration across a polarized lung epithelial monolayer that has been infected with pathogenic P. aeruginosa (PAO1). Seeding and culturing of permeable transwells with human derived lung epithelial cells is described, along with isolation of neutrophils from whole human blood and culturing of PAO1 and nonpathogenic K12 E. coli (MC1000). The emigrational process and quantitative analysis of successfully migrated neutrophils that have been mobilized in response to pathogenic infection is shown with representative data, including positive and negative controls. This in vitro model system can be manipulated and applied to other mucosal surfaces. Inflammatory responses that involve excessive neutrophil infiltration can be destructive to host tissues and can occur in the absence of pathogenic infections. A better understanding of the molecular mechanisms that promote neutrophil trans-epithelial migration through experimental manipulation of the in vitro coculture assay system described herein has significant potential to identify novel therapeutic targets for a range of mucosal infectious as well as inflammatory diseases.
Collapse
Affiliation(s)
- Mark E Kusek
- Department of Pediatrics, Harvard Medical School
| | | | | | | |
Collapse
|
88
|
Abstract
The acute respiratory distress syndrome (ARDS) is a major public health problem and a leading source of morbidity in intensive care units. Lung tissue in patients with ARDS is characterized by inflammation, with exuberant neutrophil infiltration, activation, and degranulation that is thought to initiate tissue injury through the release of proteases and oxygen radicals. Treatment of ARDS is supportive primarily because the underlying pathophysiology is poorly understood. This gap in knowledge must be addressed to identify urgently needed therapies. Recent research efforts in anti-inflammatory drug development have focused on identifying common control points in multiple signaling pathways. The protein kinase C (PKC) serine-threonine kinases are master regulators of proinflammatory signaling hubs, making them attractive therapeutic targets. Pharmacological inhibition of broad-spectrum PKC activity and, more importantly, of specific PKC isoforms (as well as deletion of PKCs in mice) exerts protective effects in various experimental models of lung injury. Furthermore, PKC isoforms have been implicated in inflammatory processes that may be involved in the pathophysiologic changes that result in ARDS, including activation of innate immune and endothelial cells, neutrophil trafficking to the lung, regulation of alveolar epithelial barrier functions, and control of neutrophil proinflammatory and prosurvival signaling. This review focuses on the mechanistic involvement of PKC isoforms in the pathogenesis of ARDS and highlights the potential of developing new therapeutic paradigms based on the selective inhibition (or activation) of specific PKC isoforms.
Collapse
|
89
|
Williams AE, Chambers RC. The mercurial nature of neutrophils: still an enigma in ARDS? Am J Physiol Lung Cell Mol Physiol 2013; 306:L217-30. [PMID: 24318116 DOI: 10.1152/ajplung.00311.2013] [Citation(s) in RCA: 287] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The acute respiratory distress syndrome (ARDS) is a life-threatening lung condition resulting from direct and indirect insults to the lung. It is characterized by disruption of the endothelial-epithelial barrier, alveolar damage, pulmonary edema, and respiratory failure. A key feature of ARDS is the accumulation of neutrophils in the lung microvasculature, interstitium, and alveolar space. Despite a clear association between neutrophil influx into the lung and disease severity, there is some debate as to whether neutrophils directly contribute to disease pathogenesis. The primary function of neutrophils is to provide immediate host defense against pathogenic microorganisms. Neutrophils release numerous antimicrobial factors such as reactive oxygen species, proteinases, and neutrophil extracellular traps. However, these factors are also toxic to host cells and can result in bystander tissue damage. The excessive accumulation of neutrophils in ARDS may therefore contribute to disease progression. Central to neutrophil recruitment is the release of chemokines, including the archetypal neutrophil chemoattractant IL-8, from resident pulmonary cells. However, the chemokine network in the inflamed lung is complex and may involve several other chemokines, including CXCL10, CCL2, and CCL7. This review will therefore focus on the experimental and clinical evidence supporting neutrophils as key players in ARDS and the chemokines involved in recruiting them into the lung.
Collapse
Affiliation(s)
- Andrew E Williams
- Centre for Inflammation and Tissue Repair, Univ. College London, Rayne Institute, 5 Univ. St., London WC1E 6JF, UK.
| | | |
Collapse
|
90
|
Kubala SA, Patil SU, Shreffler WG, Hurley BP. Pathogen induced chemo-attractant hepoxilin A3 drives neutrophils, but not eosinophils across epithelial barriers. Prostaglandins Other Lipid Mediat 2013; 108:1-8. [PMID: 24315875 DOI: 10.1016/j.prostaglandins.2013.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 11/08/2013] [Accepted: 11/21/2013] [Indexed: 12/19/2022]
Abstract
Pathogen induced migration of neutrophils across mucosal epithelial barriers requires epithelial production of the chemotactic lipid mediator, hepoxilin A3 (HXA3). HXA3 is an eicosanoid derived from arachidonic acid. Although eosinophils are also capable of penetrating mucosal surfaces, eosinophilic infiltration occurs mainly during allergic processes whereas neutrophils dominate mucosal infection. Both neutrophils and eosinophils can respond to chemotactic gradients of certain eicosanoids, however, it is not known whether eosinophils respond to pathogen induced lipid mediators such as HXA3. In this study, neutrophils and eosinophils were isolated from human blood and placed on the basolateral side of polarized epithelial monolayers grown on permeable Transwell filters and challenged by various chemotactic gradients of distinct lipid mediators. We observed that both cell populations migrated across epithelial monolayers in response to a leukotriene B4 (LTB4) gradient, whereas only eosinophils migrated toward a prostaglandin D2 (PGD2) gradient. Interestingly, while pathogen induced neutrophil trans-epithelial migration was substantial, pathogen induced eosinophil trans-epithelial migration was not observed. Further, gradients of chemotactic lipids derived from pathogen infected epithelial cells known to be enriched for HXA3 as well as purified HXA3 drove significant numbers of neutrophils across epithelial barriers, whereas eosinophils failed to respond to these gradients. These data suggest that although the eicosanoid HXA3 serves as an important neutrophil chemo-attractant at mucosal surfaces during pathogenic infection, HXA3 does not appear to exhibit chemotactic activity toward eosinophils.
Collapse
Affiliation(s)
- S A Kubala
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, United States; Center for Immunology and Inflammatory Diseases and the Food Allergy Center, Massachusetts General Hospital, Charlestown, MA 02129, United States
| | - S U Patil
- Center for Immunology and Inflammatory Diseases and the Food Allergy Center, Massachusetts General Hospital, Charlestown, MA 02129, United States
| | - W G Shreffler
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, United States; Center for Immunology and Inflammatory Diseases and the Food Allergy Center, Massachusetts General Hospital, Charlestown, MA 02129, United States
| | - B P Hurley
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, United States; Mucosal Immunology & Biology Research Center, Massachusetts General Hospital, Charlestown, MA 02129, United States.
| |
Collapse
|
91
|
Abstract
Live lung imaging has spanned the discovery of capillaries in the frog lung by Malpighi to the current use of single and multiphoton imaging of intravital and isolated perfused lung preparations incorporating fluorescent molecular probes and transgenic reporter mice. Along the way, much has been learned about the unique microcirculation of the lung, including immune cell migration and the mechanisms by which cells at the alveolar-capillary interface communicate with each other. In this review, we highlight live lung imaging techniques as applied to the role of mitochondria in lung immunity, mechanisms of signal transduction in lung compartments, studies on the composition of alveolar wall liquid, and neutrophil and platelet trafficking in the lung under homeostatic and inflammatory conditions. New applications of live lung imaging and the limitations of current techniques are discussed.
Collapse
Affiliation(s)
- Mark R. Looney
- Departments of Medicine and Laboratory Medicine, University of California, San Francisco, California 94143
| | - Jahar Bhattacharya
- Division of Pulmonary Allergy and Critical Care, Department of Medicine, and Department of Physiology & Cellular Biophysics, Columbia University College of Physicians & Surgeons, New York, New York 10032
| |
Collapse
|
92
|
Mondrinos MJ, Zhang T, Sun S, Kennedy PA, King DJ, Wolfson MR, Knight LC, Scalia R, Kilpatrick LE. Pulmonary endothelial protein kinase C-delta (PKCδ) regulates neutrophil migration in acute lung inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 184:200-13. [PMID: 24211111 DOI: 10.1016/j.ajpath.2013.09.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 08/29/2013] [Accepted: 09/23/2013] [Indexed: 12/15/2022]
Abstract
Excessive neutrophil migration across the pulmonary endothelium into the lung and release of oxidants and proteases are key elements in pathogenesis of acute lung injury. Previously, we identified protein kinase C-delta (PKCδ) as an important regulator of proinflammatory signaling in human neutrophils and demonstrated that intratracheal instillation of a TAT-conjugated PKCδ inhibitory peptide (PKCδ-TAT) is lung protective in a rat model of sepsis-induced indirect pulmonary injury (cecal ligation and puncture). In the present study, intratracheal instillation of this PKCδ inhibitor resulted in peptide distribution throughout the lung parenchyma and pulmonary endothelium and decreased neutrophil influx, with concomitant attenuation of sepsis-induced endothelial ICAM-1 and VCAM-1 expression in this model. To further delineate the role of PKCδ in regulating neutrophil migration, we used an in vitro transmigration model with human pulmonary microvascular endothelial cells (PMVECs). Consistent with in vivo findings, inhibition of PMVEC PKCδ decreased IL-1β-mediated neutrophil transmigration. PKCδ regulation was stimulus-dependent; PKCδ was required for transmigration mediated by IL-1β and fMLP (integrin-dependent), but not IL-8 (integrin-independent). PKCδ was essential for IL-1β-mediated neutrophil adherence and NF-κB-dependent expression of ICAM-1 and VCAM-1. In PMVECs, IL-1β-mediated production of ROS and activation of redox-sensitive NF-κB were PKCδ dependent, suggesting an upstream signaling role. Thus, PKCδ has an important role in regulating neutrophil-endothelial cell interactions and recruitment to the inflamed lung.
Collapse
Affiliation(s)
- Mark J Mondrinos
- Department of Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania; Center for Inflammation, Translational and Clinical Lung Research, Temple University School of Medicine, Philadelphia, Pennsylvania; Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Ting Zhang
- Department of Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania; Center for Inflammation, Translational and Clinical Lung Research, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Shuang Sun
- Department of Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania; Center for Inflammation, Translational and Clinical Lung Research, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Paul A Kennedy
- Department of Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania; Center for Inflammation, Translational and Clinical Lung Research, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Devon J King
- Department of Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania; Center for Inflammation, Translational and Clinical Lung Research, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Marla R Wolfson
- Department of Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania; Center for Inflammation, Translational and Clinical Lung Research, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Linda C Knight
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania; Department of Radiology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Rosario Scalia
- Department of Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania; Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Laurie E Kilpatrick
- Department of Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania; Center for Inflammation, Translational and Clinical Lung Research, Temple University School of Medicine, Philadelphia, Pennsylvania; Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
93
|
Costiniuk CT, Jenabian MA. The lungs as anatomical reservoirs of HIV infection. Rev Med Virol 2013; 24:35-54. [DOI: 10.1002/rmv.1772] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 09/25/2013] [Accepted: 09/26/2013] [Indexed: 12/24/2022]
Affiliation(s)
- Cecilia T. Costiniuk
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH); Durban South Africa
- Division of Infectious Diseases, Department of Medicine; University of Ottawa; Ottawa ON Canada
| | - Mohammad-Ali Jenabian
- Chronic Viral Illnesses Service; Montreal Chest Institute; Montreal QC Canada
- Research Institute; McGill University Health Centre; Montreal QC Canada
| |
Collapse
|
94
|
Bhowmick R, Maung N, Hurley BP, Ghanem EB, Gronert K, McCormick BA, Leong JM. Systemic disease during Streptococcus pneumoniae acute lung infection requires 12-lipoxygenase-dependent inflammation. THE JOURNAL OF IMMUNOLOGY 2013; 191:5115-23. [PMID: 24089193 DOI: 10.4049/jimmunol.1300522] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Acute pulmonary infection by Streptococcus pneumoniae is characterized by high bacterial numbers in the lung, a robust alveolar influx of polymorphonuclear cells (PMNs), and a risk of systemic spread of the bacterium. We investigated host mediators of S. pneumoniae-induced PMN migration and the role of inflammation in septicemia following pneumococcal lung infection. Hepoxilin A3 (HXA3) is a PMN chemoattractant and a metabolite of the 12-lipoxygenase (12-LOX) pathway. We observed that S. pneumoniae infection induced the production of 12-LOX in cultured pulmonary epithelium and in the lungs of infected mice. Inhibition of the 12-LOX pathway prevented pathogen-induced PMN transepithelial migration in vitro and dramatically reduced lung inflammation upon high-dose pulmonary challenge with S. pneumoniae in vivo, thus implicating HXA3 in pneumococcus-induced pulmonary inflammation. PMN basolateral-to-apical transmigration in vitro significantly increased apical-to-basolateral transepithelial migration of bacteria. Mice suppressed in the expression of 12-LOX exhibited little or no bacteremia and survived an otherwise lethal pulmonary challenge. Our data suggest that pneumococcal pulmonary inflammation is required for high-level bacteremia and systemic infection, partly by disrupting lung epithelium through 12-LOX-dependent HXA3 production and subsequent PMN transepithelial migration.
Collapse
Affiliation(s)
- Rudra Bhowmick
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111, USA
| | - Nang Maung
- Immune Disease Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Bryan P Hurley
- Mucosal Immunology Laboratory, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA 02129, USA
| | - Elsa Bou Ghanem
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111, USA
| | - Karsten Gronert
- Vision Science Program, School of Optometry, University of California, Berkeley, CA 94720, USA
| | - Beth A McCormick
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - John M Leong
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111, USA
| |
Collapse
|
95
|
Dixon B, Smith R, Campbell DJ, Tobin A, Newcomb AE, Rosalion A, Opeskin K, Carter H, Scott DA, Santamaria JD. The Effect of Etanercept on Lung Leukocyte Margination and Fibrin Deposition after Cardiac Surgery. Am J Respir Crit Care Med 2013; 188:751-4. [DOI: 10.1164/rccm.201301-0120le] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
96
|
Predescu DN, Bardita C, Tandon R, Predescu SA. Intersectin-1s: an important regulator of cellular and molecular pathways in lung injury. Pulm Circ 2013; 3:478-98. [PMID: 24618535 PMCID: PMC4070809 DOI: 10.1086/674439] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abstract Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are severe syndromes resulting from the diffuse damage of the pulmonary parenchyma. ALI and ARDS are induced by a plethora of local or systemic insults, leading to the activation of multiple pathways responsible for injury, resolution, and repair or scarring of the lungs. Despite the large efforts aimed at exploring the roles of different pathways in humans and animal models and the great strides made in understanding the pathogenesis of ALI/ARDS, the only viable treatment options are still dependent on ventilator and cardiovascular support. Investigation of the pathophysiological mechanisms responsible for initiation and resolution or advancement toward lung scarring in ALI/ARDS animal models led to a better understanding of the disease's complexity and helped in elucidating the links between ALI and systemic multiorgan failure. Although animal models of ALI/ARDS have pointed out a variety of new ideas for study, there are still limited data regarding the initiating factors, the critical steps in the progression of the disease, and the central mechanisms dictating its resolution or progression to lung scarring. Recent studies link deficiency of intersectin-1s (ITSN-1s), a prosurvival protein of lung endothelial cells, to endothelial barrier dysfunction and pulmonary edema as well as to the repair/recovery from ALI. This review discusses the effects of ITSN-1s deficiency on pulmonary endothelium and its significance in the pathology of ALI/ARDS.
Collapse
Affiliation(s)
- Dan N Predescu
- 1 Department of Pharmacology, Rush University, Chicago, Illinois, USA
| | | | | | | |
Collapse
|
97
|
Van Ness M, Jensen H, Adamson GN, Kysar PE, Holland P. Neutrophils contain cholesterol crystals in transfusion-related acute lung injury (TRALI). Am J Clin Pathol 2013; 140:170-6. [PMID: 23897251 DOI: 10.1309/ajcpgkojv15avznt] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
OBJECTIVES Intracellular components of transfusion-related acute lung injury (TRALI) were investigated by transmission electron microscopy. METHODS The lungs from 2 fatal TRALI cases and 2 controls, previously studied by scanning electron microscopy, were studied by transmission electron microscopy. Morphologic data by light and phase microscopy, along with scanning and transmission electron microscopic observations, were collated. RESULTS The 2 fatal TRALI cases exhibited dense laminated material within capillaries and postcapillary venules, similar to material identified within their neutrophils when viewed by transmission electron microscopy. This material polarized light and is presumed to be cholesterol crystals. CONCLUSIONS The damage to the pulmonary vascular endothelium in TRALI is related to formation of cholesterol crystals originating within neutrophils.
Collapse
Affiliation(s)
- Michael Van Ness
- Department of Pathology and Laboratory Medicine, University of California Davis Health System, Sacramento, CA
| | - Hanne Jensen
- Department of Pathology and Laboratory Medicine, University of California Davis Health System, Sacramento, CA
| | - Grete N. Adamson
- Department of Pathology and Laboratory Medicine, University of California Davis Health System, Sacramento, CA
| | - Patricia E. Kysar
- Department of Pathology and Laboratory Medicine, University of California Davis Health System, Sacramento, CA
| | - Paul Holland
- Department of Pathology and Laboratory Medicine, University of California Davis Health System, Sacramento, CA
| |
Collapse
|
98
|
Raju SV, Painter RG, Bagby GJ, Nelson S, Wang G. Response of Differentiated Human Airway Epithelia to Alcohol Exposure and Klebsiella Pneumoniae Challenge. Med Sci (Basel) 2013; 1:2-19. [PMID: 25485141 PMCID: PMC4255281 DOI: 10.3390/medsci1010002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Alcohol abuse has been associated with increased susceptibility to pulmonary infection. It is not fully defined how alcohol contributes to the host defense compromise. Here primary human airway epithelial cells were cultured at an air-liquid interface to form a differentiated and polarized epithelium. This unique culture model allowed us to closely mimic lung infection in the context of alcohol abuse by basolateral alcohol exposure and apical live bacterial challenge. Application of clinically relevant concentrations of alcohol for 24 hours did not significantly alter epithelial integrity or barrier function. When apically challenged with viable Klebsiella pneumoniae, the cultured epithelia had an enhanced tightness which was unaffected by alcohol. Further, alcohol enhanced apical bacterial growth, but not bacterial binding to the cells. The cultured epithelium in the absence of any treatment or stimulation had a base-level IL-6 and IL-8 secretion. Apical bacterial challenge significantly elevated the basolateral secretion of inflammatory cytokines including IL-2, IL-4, IL-6, IL-8, IFN-γ, GM-CSF, and TNF-α. However, alcohol suppressed the observed cytokine burst in response to infection. Addition of adenosine receptor agonists negated the suppression of IL-6 and TNF-α. Thus, acute alcohol alters the epithelial cytokine response to infection, which can be partially mitigated by adenosine receptor agonists.
Collapse
Affiliation(s)
- Sammeta V. Raju
- Comprehensive Alcohol Research Center, Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Richard G. Painter
- Comprehensive Alcohol Research Center, Department of Microbiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Gregory J. Bagby
- Comprehensive Alcohol Research Center, Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Steve Nelson
- Comprehensive Alcohol Research Center, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Guoshun Wang
- Comprehensive Alcohol Research Center, Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Comprehensive Alcohol Research Center, Department of Microbiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Comprehensive Alcohol Research Center, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
99
|
Hoenderdos K, Condliffe A. The Neutrophil in Chronic Obstructive Pulmonary Disease. Too Little, Too Late or Too Much, Too Soon? Am J Respir Cell Mol Biol 2013; 48:531-9. [DOI: 10.1165/rcmb.2012-0492tr] [Citation(s) in RCA: 260] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
100
|
Transendothelial migration enables subsequent transmigration of neutrophils through underlying pericytes. PLoS One 2013; 8:e60025. [PMID: 23555870 PMCID: PMC3608600 DOI: 10.1371/journal.pone.0060025] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 02/21/2013] [Indexed: 01/13/2023] Open
Abstract
During acute inflammation, neutrophil recruitment into extravascular tissue requires neutrophil tethering and rolling on cytokine-activated endothelial cells (ECs), tight adhesion, crawling towards EC junctions and transendothelial migration (TEM). Following TEM, neutrophils must still traverse the subendothelial basement membrane and network of pericytes (PCs). Until recently, the contribution of the PC layer to neutrophil recruitment was largely ignored. Here we analyze human neutrophil interactions with interleukin (IL)-1β-activated human EC monolayers, PC monolayers and EC/PC bilayers in vitro. Compared to EC, PC support much lower levels of neutrophil binding (54.6% vs. 7.1%, respectively) and transmigration (63.7 vs. 8.8%, respectively) despite comparable levels of IL-8 (CXCL8) synthesis and display. Remarkably, EC/PC bilayers support intermediate levels of transmigration (37.7%). Neutrophil adhesion to both cell types is Mac-1-dependent and while ICAM-1 transduction of PCs increases neutrophil adhesion to (41.4%), it does not increase transmigration through PC monolayers. TEM, which increases neutrophil Mac-1 surface expression, concomitantly increases the ability of neutrophils to traverse PCs (19.2%). These data indicate that contributions from both PCs and ECs must be considered in evaluation of microvasculature function in acute inflammation.
Collapse
|