Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lin L. Mantell is active.

Publication


Featured researches published by Lin L. Mantell.


Journal of Immunotoxicology | 2010

Hyperoxia sensing: From molecular mechanisms to significance in disease

Ashwini Gore; Maitreyi Muralidhar; Michael Graham Espey; Kurt Degenhardt; Lin L. Mantell

Oxygen therapy using mechanical ventilation with hyperoxia is necessary to treat patients with respiratory failure and distress. However, prolonged exposure to hyperoxia leads to the generation of excessive reactive oxygen species (ROS), causing cellular damage and multiple organ dysfunctions. As the lungs are directly exposed, hyperoxia can cause both acute and chronic inflammatory lung injury and compromise innate immunity. ROS may contribute to pulmonary oxygen toxicity by overwhelming redox homeostasis, altering signaling cascades that affect cell fate, ultimately leading to hyperoxia-induced acute lung injury (HALI). HALI is characterized by pronounced inflammatory responses with leukocyte infiltration, injury, and death of pulmonary cells, including epithelia, endothelia, and macrophages. Under hyperoxic conditions, ROS mediate both direct and indirect modulation of signaling molecules such as protein kinases, transcription factors, receptors, and pro- and anti-apoptotic factors. The focus of this review is to elaborate on hyperoxia-activated key sensing molecules and current understanding of their signaling mechanisms in HALI. A better understanding of the signaling pathways leading to HALI may provide valuable insights on its pathogenesis and may help in designing more effective therapeutic approaches.


American Journal of Respiratory Cell and Molecular Biology | 2013

High Mobility Group Box–1 Mediates Hyperoxia-Induced Impairment of Pseudomonas aeruginosa Clearance and Inflammatory Lung Injury in Mice

Vivek Patel; Ravikumar Sitapara; Ashwini Gore; Binh Phan; Lokesh Sharma; Vaishali Sampat; Jianhua Li; Huan Yang; Sangeeta Chavan; Haichao Wang; Kevin J. Tracey; Lin L. Mantell

Mechanical ventilation with supraphysiological concentrations of oxygen (hyperoxia) is routinely used to treat patients with respiratory distress. However, a significant number of patients on ventilators exhibit enhanced susceptibility to infections and develop ventilator-associated pneumonia (VAP). Pseudomonas aeruginosa (PA) is one of the most common species of bacteria found in these patients. Previously, we demonstrated that prolonged exposure to hyperoxia can compromise the ability of alveolar macrophages (AMs), an essential part of the innate immunity, to phagocytose PA. This study sought to investigate the potential molecular mechanisms underlying hyperoxia-compromised innate immunity against bacterial infection in a murine model of PA pneumonia. Here, we show that exposure to hyperoxia (≥ 99% O2) led to a significant elevation in concentrations of airway high mobility group box-1 (HMGB1) and increased mortality in C57BL/6 mice infected with PA. Treatment of these mice with a neutralizing anti-HMGB1 monoclonal antibody (mAb) resulted in a reduction in bacterial counts, injury, and numbers of neutrophils in the lungs, and an increase in leukocyte phagocytic activity compared with mice receiving control mAb. This improved phagocytic function was associated with reduced concentrations of airway HMGB1. The correlation between phagocytic activity and concentrations of extracellular HMGB1 was also observed in cultured macrophages. These results indicate a pathogenic role for HMGB1 in hyperoxia-induced impairment with regard to a hosts ability to clear bacteria and inflammatory lung injury. Thus, HMGB1 may provide a novel molecular target for improving hyperoxia-compromised innate immunity in patients with VAP.


Redox biology | 2014

Inhibition of extracellular HMGB1 attenuates hyperoxia-induced inflammatory acute lung injury.

Maria Entezari; Mohammad Javdan; Daniel J. Antoine; Dympna M.P. Morrow; Ravikumar Sitapara; Vivek Patel; Mao Wang; Lokesh Sharma; Samir Gorasiya; Michelle Zur; Wenjun Wu; Jian Hua Li; Huan Yang; Charles R. Ashby; Douglas D. Thomas; Haichao Wang; Lin L. Mantell

Prolonged exposure to hyperoxia results in acute lung injury (ALI), accompanied by a significant elevation in the levels of proinflammatory cytokines and leukocyte infiltration in the lungs. However, the mechanisms underlying hyperoxia-induced proinflammatory ALI remain to be elucidated. In this study, we investigated the role of the proinflammatory cytokine high mobility group box protein 1 (HMGB1) in hyperoxic inflammatory lung injury, using an adult mouse model. The exposure of C57BL/6 mice to ≥99% O2 (hyperoxia) significantly increased the accumulation of HMGB1 in the bronchoalveolar lavage fluids (BALF) prior to the onset of severe inflammatory lung injury. In the airways of hyperoxic mice, HMGB1 was hyperacetylated and existed in various redox forms. Intratracheal administration of recombinant HMGB1 (rHMGB1) caused a significant increase in leukocyte infiltration into the lungs compared to animal treated with a non-specific peptide. Neutralizing anti-HMGB1 antibodies, administrated before hyperoxia significantly attenuated pulmonary edema and inflammatory responses, as indicated by decreased total protein content, wet/dry weight ratio, and numbers of leukocytes in the airways. This protection was also observed when HMGB1 inhibitors were administered after the onset of the hyperoxic exposure. The aliphatic antioxidant, ethyl pyruvate (EP), inhibited HMGB1 secretion from hyperoxic macrophages and attenuated hyperoxic lung injury. Overall, our data suggest that HMGB1 plays a critical role in mediating hyperoxic ALI through the recruitment of leukocytes into the lungs. If these results can be translated to humans, they suggest that HMGB1 inhibitors provide treatment regimens for oxidative inflammatory lung injury in patients receiving hyperoxia through mechanical ventilation.


Molecular Medicine | 2014

The α7 Nicotinic Acetylcholine Receptor Agonist GTS-21 Improves Bacterial Clearance in Mice by Restoring Hyperoxia-Compromised Macrophage Function

Ravikumar Sitapara; Daniel J. Antoine; Lokesh Sharma; Vivek Patel; Charles R. Ashby; Samir Gorasiya; Huan Yang; Michelle Zur; Lin L. Mantell

Mechanical ventilation with supraphysiological concentrations of oxygen (hyperoxia) is routinely used to treat patients with respiratory distress. However, prolonged exposure to hyperoxia compromises the ability of the macrophage to phagocytose and clear bacteria. Previously, we showed that the exposure of mice to hyperoxia elicits the release of the nuclear protein high mobility group box-1 (HMGB1) into the airways. Extracellular HMGB1 impairs macrophage phagocytosis and increases the mortality of mice infected with Pseudomonas aeruginosa (PA). The aim of this study was to determine whether GTS-21 [3-(2,4 dimethoxy-benzylidene)-anabaseine dihydrochloride], an α7 nicotinic acetylcholine receptor (α7nAChR) agonist, could inhibit hyperoxia-in-duced HMGB1 release into the airways, enhance macrophage function and improve bacterial clearance from the lungs in a mouse model of ventilator-associated pneumonia. GTS-21 (0.04, 0.4 and 4 mg/kg) or saline was systemically administered via intraperitoneal injection to mice that were exposed to hyperoxia (≥99% O2) and subsequently challenged with PA. We found that systemic administration of 4 mg/kg GTS-21 significantly increased bacterial clearance, decreased acute lung injury and decreased accumulation of airway HMGB1. To investigate the cellular mechanism of these observations, RAW 264.7 cells, a macrophagelike cell line, were incubated with different concentrations of GTS-21 in the presence of 95% O2. The phagocytic activity of macrophages was significantly increased by GTS-21 in a dose-dependent manner. In addition, hyperoxia-induced hyperacetylation of HMGB1 was significantly reduced in macrophages incubated with GTS-21. Furthermore, GTS-21 significantly inhibited the cytoplasmic translocation and release of HMGB1 from these macrophages. Our results indicate that GTS-21 is effective in improving bacterial clearance and reducing acute lung injury by enhancing macrophage function via inhibiting the release of nuclear HMGB1. Therefore, the α7nAChR represents a possible pharmacological target to improve the clinical outcome of patients on ventilators by augmenting host defense against bacterial infections.


Cancer Research | 2015

Nitric Oxide Regulates Gene Expression in Cancers by Controlling Histone Posttranslational Modifications

Divya Vasudevan; Jason R. Hickok; Rhea C. Bovee; Vy T. Pham; Lin L. Mantell; Neil Bahroos; Pinal Kanabar; Xing Jun Cao; Mark Maienschein-Cline; Benjamin A. Garcia; Douglas D. Thomas

Altered nitric oxide (•NO) metabolism underlies cancer pathology, but mechanisms explaining many •NO-associated phenotypes remain unclear. We have found that cellular exposure to •NO changes histone posttranslational modifications (PTM) by directly inhibiting the catalytic activity of JmjC-domain containing histone demethylases. Herein, we describe how •NO exposure links modulation of histone PTMs to gene expression changes that promote oncogenesis. Through high-resolution mass spectrometry, we generated an extensive map of •NO-mediated histone PTM changes at 15 critical lysine residues on the core histones H3 and H4. Concomitant microarray analysis demonstrated that exposure to physiologic •NO resulted in the differential expression of over 6,500 genes in breast cancer cells. Measurements of the association of H3K9me2 and H3K9ac across genomic loci revealed that differential distribution of these particular PTMs correlated with changes in the level of expression of numerous oncogenes, consistent with epigenetic code. Our results establish that •NO functions as an epigenetic regulator of gene expression mediated by changes in histone PTMs.


Methods of Molecular Biology | 2014

Assessment of Phagocytic Activity of Cultured Macrophages Using Fluorescence Microscopy and Flow Cytometry

Lokesh Sharma; Wenjun Wu; Sanjay L. Dholakiya; Samir Gorasiya; Jiao Wu; Ravikumar Sitapara; Vivek Patel; Mao Wang; Michelle Zur; Shloka Reddy; Nathan Siegelaub; Katrina Bamba; Frank A. Barile; Lin L. Mantell

Phagocytosis is the process by which phagocytes, including macrophages, neutrophils and monocytes, engulf and kill invading pathogens, remove foreign particles, and clear cell debris. Phagocytes and their ability to phagocytose are an important part of the innate immune system and are critical for homeostasis of the host. Impairment in phagocytosis has been associated with numerous diseases and disorders. Different cytokines have been shown to affect the phagocytic process. Cytokines including TNFα, IL-1β, GM-CSF, and TGF-β1 were found to promote phagocytosis, whereas high mobility group box-1 (HMGB1) inhibited the phagocytic function of macrophages. Here, we describe two commonly used methods to assess the phagocytic function of cultured macrophages, which can easily be applied to other phagocytes. Each method is based on the extent of engulfment of FITC-labeled latex minibeads by macrophages under different conditions. Phagocytic activity can be assessed either by counting individual cells using a fluorescence microscope or measuring fluorescence intensity using a flow cytometer.


Journal of Immunotoxicology | 2011

Hydrogen peroxide enhances phagocytosis of Pseudomonas aeruginosa in hyperoxia

Binh Phan; Maria Entezari; Richard A. Lockshin; Diana C. Bartelt; Lin L. Mantell

Mechanical ventilation with hyperoxia is a necessary treatment for patients with respiratory distress. However, patients on mechanical ventilation have increased susceptibility to infection. Studies including ours have shown that reactive oxygen species (ROS), generated by exposure to prolonged hyperoxia, can cause a decrease in the phagocytic activity of alveolar macrophages. Hydrogen peroxide (H2O2) is a form of ROS generated under hyperoxic conditions. In this study, we examined whether treatment with H2O2 directly affects macrophage phagocytic ability in RAW 264.7 cells that were exposed to either 21% O2 (room air) or 95% O2 (hyperoxia). Moderate concentrations (ranging from 10 to 250 µM) of H2O2 significantly enhanced macrophage phagocytic activity and restored hyperoxia-suppressed phagocytosis through attenuation of hyperoxia-induced disorganization of actin cytoskeleton and actin oxidation. These results indicate that H2O2 at low–moderate concentrations can be beneficial to host immune responses by improving macrophage phagocytic activity.


American Journal of Respiratory Cell and Molecular Biology | 2000

Interleukin-6–Induced Protection in Hyperoxic Acute Lung Injury

Nicholas S. Ward; Aaron B. Waxman; Robert J. Homer; Lin L. Mantell; Oskar Einarsson; YuFen Du; Jack A. Elias


Free Radical Biology and Medicine | 2007

Hyperoxia-induced signal transduction pathways in pulmonary epithelial cells

Tahereh E. Zaher; Edmund J. Miller; Dympna M. P. Morrow; Mohammad Javdan; Lin L. Mantell


American Journal of Respiratory Cell and Molecular Biology | 2003

Interleukin-11 and Interleukin-6 Protect Cultured Human Endothelial Cells from H2O2-Induced Cell Death

Aaron B. Waxman; Keyvan Mahboubi; Roy G. Knickelbein; Lin L. Mantell; Nicholas Manzo; Jordan S. Pober; Jack A. Elias

Collaboration


Dive into the Lin L. Mantell's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Douglas D. Thomas

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Haichao Wang

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mao Wang

St. John's University

View shared research outputs
Researchain Logo
Decentralizing Knowledge