198 results on '"Rousseau, Simon"'
Search Results
152. Clinical utilization of genomics data produced by the international Pseudomonas aeruginosa consortium
- Author
-
Freschi, Luca, Jeukens, Julie, Kukavica-Ibrulj, Irena, Boyle, Brian, Maaroufi, Halim, Fothergill, Joanne L., Moore, Matthew, Winsor, Geoffrey L., Aaron, Shawn D., Barbeau, Jean, Bell, Scott C., Burns, Jane L., Charette, Steve J., Dewar, Ken, Grimwood, Keith, Hancock, Robert E.W., Harrison, Joe J., Heeb, Stephan, Jelsbak, Lars, Jia, Baofeng, Kenna, Dervla T., Kidd, Timothy J., Klockgether, Jens, Lam, Joseph S., Lamont, Iain L., Lewenza, Shawn, Loman, Nick, Manos, Jim, McArthur, Andrew G., McKeown, Josie, Milot, Julie, Naghra, Hardeep, Nguyen, Dao, Pereira, Sheldon K., Perron, Gabriel G., Pirnay, Jean-Paul, Rainey, Paul B., Rousseau, Simon, Santos, Pedro M., Stephenson, Anne, Turton, Jane F., Waglechner, Nicholas, Williams, Paul, Thrane, Sandra W., Wright, Gerard D., Brinkman, Fiona S.L., Tucker, Nicholas P., Winstanley, Craig, and Levesque, Roger C.
- Subjects
Pseudomonas aeruginosa, next-generation sequencing, bacterial genome, phylogeny, database,cystic fibrosis, antibiotic resistance, clinical microbiology - Abstract
The International Pseudomonas aeruginosa Consortium is sequencing over 1000 genomes and building an analysis pipeline for the study of Pseudomonas genome evolution, antibiotic resistance and virulence genes. Metadata, including genomic and phenotypic data for each isolate of the collection, are available through the International Pseudomonas Consortium Database (http://ipcd.ibis.ulaval.ca/). Here, we present our strategy and the results that emerged from the analysis of the first 389 genomes. With as yet unmatched resolution, our results confirm that P. aeruginosa strains can be divided into three major groups that are further divided into subgroups, some not previously reported in the literature. We also provide the first snapshot of P. aeruginosa strain diversity with respect to antibiotic resistance. Our approach will allow us to draw potential links between environmental strains and those implicated in human and animal infections, understand how patients become infected and how the infection evolves over time as well as identify prognostic markers for better evidence-based decisions on patient care.
153. Clinical utilization of genomics data produced by the international Pseudomonas aeruginosa consortium
- Author
-
Freschi, Luca, Jeukens, Julie, Kukavica-Ibrulj, Irena, Boyle, Brian, Dupont, Marie-Josée, Laroche, Jérôme, Larose, Stéphane, Maaroufi, Halim, Fothergill, Joanne L., Moore, Matthew, Winsor, Geoffrey L., Aaron, Shawn D., Barbeau, Jean, Bell, Scott C., Burns, Jane L., Cámara, Miguel, Cantin, André, Charette, Steve J., Dewar, Ken, Déziel, Éric, Grimwood, Keith, Hancock, Robert E.W., Harrison, Joe J., Heeb, Stephan, Jelsbak, Lars, Jia, Baofeng, Kenna, Dervla T., Kidd, Timothy J., Klockgether, Jens, Lam, Joseph S., Lamont, Iain L., Lewenza, Shawn, Loman, Nick, Malouin, François, Manos, Jim, McArthur, Andrew G., McKeown, Josie, Milot, Julie, Naghra, Hardeep, Nguyen, Dao, Pereira, Sheldon K., Perron, Gabriel G., Pirnay, Jean-Paul, Rainey, Paul B., Rousseau, Simon, Santos, Pedro M., Stephenson, Anne, Taylor, Véronique, Turton, Jane F., Waglechner, Nicholas, Williams, Paul, Thrane, Sandra W., Wright, Gerard D., Brinkman, Fiona S.L., Tucker, Nicholas P., Tümmler, Burkhard, Winstanley, Craig, Levesque, Roger C., Freschi, Luca, Jeukens, Julie, Kukavica-Ibrulj, Irena, Boyle, Brian, Dupont, Marie-Josée, Laroche, Jérôme, Larose, Stéphane, Maaroufi, Halim, Fothergill, Joanne L., Moore, Matthew, Winsor, Geoffrey L., Aaron, Shawn D., Barbeau, Jean, Bell, Scott C., Burns, Jane L., Cámara, Miguel, Cantin, André, Charette, Steve J., Dewar, Ken, Déziel, Éric, Grimwood, Keith, Hancock, Robert E.W., Harrison, Joe J., Heeb, Stephan, Jelsbak, Lars, Jia, Baofeng, Kenna, Dervla T., Kidd, Timothy J., Klockgether, Jens, Lam, Joseph S., Lamont, Iain L., Lewenza, Shawn, Loman, Nick, Malouin, François, Manos, Jim, McArthur, Andrew G., McKeown, Josie, Milot, Julie, Naghra, Hardeep, Nguyen, Dao, Pereira, Sheldon K., Perron, Gabriel G., Pirnay, Jean-Paul, Rainey, Paul B., Rousseau, Simon, Santos, Pedro M., Stephenson, Anne, Taylor, Véronique, Turton, Jane F., Waglechner, Nicholas, Williams, Paul, Thrane, Sandra W., Wright, Gerard D., Brinkman, Fiona S.L., Tucker, Nicholas P., Tümmler, Burkhard, Winstanley, Craig, and Levesque, Roger C.
- Abstract
The International Pseudomonas aeruginosa Consortium is sequencing over 1000 genomes and building an analysis pipeline for the study of Pseudomonas genome evolution, antibiotic resistance and virulence genes. Metadata, including genomic and phenotypic data for each isolate of the collection, are available through the International Pseudomonas Consortium Database (http://ipcd.ibis.ulaval.ca/). Here, we present our strategy and the results that emerged from the analysis of the first 389 genomes. With as yet unmatched resolution, our results confirm that P. aeruginosa strains can be divided into three major groups that are further divided into subgroups, some not previously reported in the literature. We also provide the first snapshot of P. aeruginosa strain diversity with respect to antibiotic resistance. Our approach will allow us to draw potential links between environmental strains and those implicated in human and animal infections, understand how patients become infected and how the infection evolves over time as well as identify prognostic markers for better evidence-based decisions on patient care.
- Full Text
- View/download PDF
154. Clinical utilization of genomics data produced by the international Pseudomonas aeruginosa consortium
- Author
-
Freschi, Luca, Jeukens, Julie, Kukavica-Ibrulj, Irena, Boyle, Brian, Dupont, Marie-Josée, Laroche, Jérôme, Larose, Stéphane, Maaroufi, Halim, Fothergill, Joanne L., Moore, Matthew, Winsor, Geoffrey L., Aaron, Shawn D., Barbeau, Jean, Bell, Scott C., Burns, Jane L., Cámara, Miguel, Cantin, André, Charette, Steve J., Dewar, Ken, Déziel, Éric, Grimwood, Keith, Hancock, Robert E.W., Harrison, Joe J., Heeb, Stephan, Jelsbak, Lars, Jia, Baofeng, Kenna, Dervla T., Kidd, Timothy J., Klockgether, Jens, Lam, Joseph S., Lamont, Iain L., Lewenza, Shawn, Loman, Nick, Malouin, François, Manos, Jim, McArthur, Andrew G., McKeown, Josie, Milot, Julie, Naghra, Hardeep, Nguyen, Dao, Pereira, Sheldon K., Perron, Gabriel G., Pirnay, Jean-Paul, Rainey, Paul B., Rousseau, Simon, Santos, Pedro M., Stephenson, Anne, Taylor, Véronique, Turton, Jane F., Waglechner, Nicholas, Williams, Paul, Thrane, Sandra W., Wright, Gerard D., Brinkman, Fiona S.L., Tucker, Nicholas P., Tümmler, Burkhard, Winstanley, Craig, Levesque, Roger C., Freschi, Luca, Jeukens, Julie, Kukavica-Ibrulj, Irena, Boyle, Brian, Dupont, Marie-Josée, Laroche, Jérôme, Larose, Stéphane, Maaroufi, Halim, Fothergill, Joanne L., Moore, Matthew, Winsor, Geoffrey L., Aaron, Shawn D., Barbeau, Jean, Bell, Scott C., Burns, Jane L., Cámara, Miguel, Cantin, André, Charette, Steve J., Dewar, Ken, Déziel, Éric, Grimwood, Keith, Hancock, Robert E.W., Harrison, Joe J., Heeb, Stephan, Jelsbak, Lars, Jia, Baofeng, Kenna, Dervla T., Kidd, Timothy J., Klockgether, Jens, Lam, Joseph S., Lamont, Iain L., Lewenza, Shawn, Loman, Nick, Malouin, François, Manos, Jim, McArthur, Andrew G., McKeown, Josie, Milot, Julie, Naghra, Hardeep, Nguyen, Dao, Pereira, Sheldon K., Perron, Gabriel G., Pirnay, Jean-Paul, Rainey, Paul B., Rousseau, Simon, Santos, Pedro M., Stephenson, Anne, Taylor, Véronique, Turton, Jane F., Waglechner, Nicholas, Williams, Paul, Thrane, Sandra W., Wright, Gerard D., Brinkman, Fiona S.L., Tucker, Nicholas P., Tümmler, Burkhard, Winstanley, Craig, and Levesque, Roger C.
- Abstract
The International Pseudomonas aeruginosa Consortium is sequencing over 1000 genomes and building an analysis pipeline for the study of Pseudomonas genome evolution, antibiotic resistance and virulence genes. Metadata, including genomic and phenotypic data for each isolate of the collection, are available through the International Pseudomonas Consortium Database (http://ipcd.ibis.ulaval.ca/). Here, we present our strategy and the results that emerged from the analysis of the first 389 genomes. With as yet unmatched resolution, our results confirm that P. aeruginosa strains can be divided into three major groups that are further divided into subgroups, some not previously reported in the literature. We also provide the first snapshot of P. aeruginosa strain diversity with respect to antibiotic resistance. Our approach will allow us to draw potential links between environmental strains and those implicated in human and animal infections, understand how patients become infected and how the infection evolves over time as well as identify prognostic markers for better evidence-based decisions on patient care.
- Full Text
- View/download PDF
155. Clinical utilization of genomics data produced by the international Pseudomonas aeruginosa consortium
- Author
-
Freschi, Luca, Jeukens, Julie, Kukavica-Ibrulj, Irena, Boyle, Brian, Dupont, Marie-Josée, Laroche, Jérôme, Larose, Stéphane, Maaroufi, Halim, Fothergill, Joanne L., Moore, Matthew, Winsor, Geoffrey L., Aaron, Shawn D., Barbeau, Jean, Bell, Scott C., Burns, Jane L., Cámara, Miguel, Cantin, André, Charette, Steve J., Dewar, Ken, Déziel, Éric, Grimwood, Keith, Hancock, Robert E.W., Harrison, Joe J., Heeb, Stephan, Jelsbak, Lars, Jia, Baofeng, Kenna, Dervla T., Kidd, Timothy J., Klockgether, Jens, Lam, Joseph S., Lamont, Iain L., Lewenza, Shawn, Loman, Nick, Malouin, François, Manos, Jim, McArthur, Andrew G., McKeown, Josie, Milot, Julie, Naghra, Hardeep, Nguyen, Dao, Pereira, Sheldon K., Perron, Gabriel G., Pirnay, Jean-Paul, Rainey, Paul B., Rousseau, Simon, Santos, Pedro M., Stephenson, Anne, Taylor, Véronique, Turton, Jane F., Waglechner, Nicholas, Williams, Paul, Thrane, Sandra W., Wright, Gerard D., Brinkman, Fiona S.L., Tucker, Nicholas P., Tümmler, Burkhard, Winstanley, Craig, Levesque, Roger C., Freschi, Luca, Jeukens, Julie, Kukavica-Ibrulj, Irena, Boyle, Brian, Dupont, Marie-Josée, Laroche, Jérôme, Larose, Stéphane, Maaroufi, Halim, Fothergill, Joanne L., Moore, Matthew, Winsor, Geoffrey L., Aaron, Shawn D., Barbeau, Jean, Bell, Scott C., Burns, Jane L., Cámara, Miguel, Cantin, André, Charette, Steve J., Dewar, Ken, Déziel, Éric, Grimwood, Keith, Hancock, Robert E.W., Harrison, Joe J., Heeb, Stephan, Jelsbak, Lars, Jia, Baofeng, Kenna, Dervla T., Kidd, Timothy J., Klockgether, Jens, Lam, Joseph S., Lamont, Iain L., Lewenza, Shawn, Loman, Nick, Malouin, François, Manos, Jim, McArthur, Andrew G., McKeown, Josie, Milot, Julie, Naghra, Hardeep, Nguyen, Dao, Pereira, Sheldon K., Perron, Gabriel G., Pirnay, Jean-Paul, Rainey, Paul B., Rousseau, Simon, Santos, Pedro M., Stephenson, Anne, Taylor, Véronique, Turton, Jane F., Waglechner, Nicholas, Williams, Paul, Thrane, Sandra W., Wright, Gerard D., Brinkman, Fiona S.L., Tucker, Nicholas P., Tümmler, Burkhard, Winstanley, Craig, and Levesque, Roger C.
- Abstract
The International Pseudomonas aeruginosa Consortium is sequencing over 1000 genomes and building an analysis pipeline for the study of Pseudomonas genome evolution, antibiotic resistance and virulence genes. Metadata, including genomic and phenotypic data for each isolate of the collection, are available through the International Pseudomonas Consortium Database (http://ipcd.ibis.ulaval.ca/). Here, we present our strategy and the results that emerged from the analysis of the first 389 genomes. With as yet unmatched resolution, our results confirm that P. aeruginosa strains can be divided into three major groups that are further divided into subgroups, some not previously reported in the literature. We also provide the first snapshot of P. aeruginosa strain diversity with respect to antibiotic resistance. Our approach will allow us to draw potential links between environmental strains and those implicated in human and animal infections, understand how patients become infected and how the infection evolves over time as well as identify prognostic markers for better evidence-based decisions on patient care.
- Full Text
- View/download PDF
156. Clinical utilization of genomics data produced by the international Pseudomonas aeruginosa consortium
- Author
-
Freschi, Luca, Jeukens, Julie, Kukavica-Ibrulj, Irena, Boyle, Brian, Dupont, Marie-Josée, Laroche, Jérôme, Larose, Stéphane, Maaroufi, Halim, Fothergill, Joanne L., Moore, Matthew, Winsor, Geoffrey L., Aaron, Shawn D., Barbeau, Jean, Bell, Scott C., Burns, Jane L., Cámara, Miguel, Cantin, André, Charette, Steve J., Dewar, Ken, Déziel, Éric, Grimwood, Keith, Hancock, Robert E.W., Harrison, Joe J., Heeb, Stephan, Jelsbak, Lars, Jia, Baofeng, Kenna, Dervla T., Kidd, Timothy J., Klockgether, Jens, Lam, Joseph S., Lamont, Iain L., Lewenza, Shawn, Loman, Nick, Malouin, François, Manos, Jim, McArthur, Andrew G., McKeown, Josie, Milot, Julie, Naghra, Hardeep, Nguyen, Dao, Pereira, Sheldon K., Perron, Gabriel G., Pirnay, Jean-Paul, Rainey, Paul B., Rousseau, Simon, Santos, Pedro M., Stephenson, Anne, Taylor, Véronique, Turton, Jane F., Waglechner, Nicholas, Williams, Paul, Thrane, Sandra W., Wright, Gerard D., Brinkman, Fiona S.L., Tucker, Nicholas P., Tümmler, Burkhard, Winstanley, Craig, Levesque, Roger C., Freschi, Luca, Jeukens, Julie, Kukavica-Ibrulj, Irena, Boyle, Brian, Dupont, Marie-Josée, Laroche, Jérôme, Larose, Stéphane, Maaroufi, Halim, Fothergill, Joanne L., Moore, Matthew, Winsor, Geoffrey L., Aaron, Shawn D., Barbeau, Jean, Bell, Scott C., Burns, Jane L., Cámara, Miguel, Cantin, André, Charette, Steve J., Dewar, Ken, Déziel, Éric, Grimwood, Keith, Hancock, Robert E.W., Harrison, Joe J., Heeb, Stephan, Jelsbak, Lars, Jia, Baofeng, Kenna, Dervla T., Kidd, Timothy J., Klockgether, Jens, Lam, Joseph S., Lamont, Iain L., Lewenza, Shawn, Loman, Nick, Malouin, François, Manos, Jim, McArthur, Andrew G., McKeown, Josie, Milot, Julie, Naghra, Hardeep, Nguyen, Dao, Pereira, Sheldon K., Perron, Gabriel G., Pirnay, Jean-Paul, Rainey, Paul B., Rousseau, Simon, Santos, Pedro M., Stephenson, Anne, Taylor, Véronique, Turton, Jane F., Waglechner, Nicholas, Williams, Paul, Thrane, Sandra W., Wright, Gerard D., Brinkman, Fiona S.L., Tucker, Nicholas P., Tümmler, Burkhard, Winstanley, Craig, and Levesque, Roger C.
- Abstract
The International Pseudomonas aeruginosa Consortium is sequencing over 1000 genomes and building an analysis pipeline for the study of Pseudomonas genome evolution, antibiotic resistance and virulence genes. Metadata, including genomic and phenotypic data for each isolate of the collection, are available through the International Pseudomonas Consortium Database (http://ipcd.ibis.ulaval.ca/). Here, we present our strategy and the results that emerged from the analysis of the first 389 genomes. With as yet unmatched resolution, our results confirm that P. aeruginosa strains can be divided into three major groups that are further divided into subgroups, some not previously reported in the literature. We also provide the first snapshot of P. aeruginosa strain diversity with respect to antibiotic resistance. Our approach will allow us to draw potential links between environmental strains and those implicated in human and animal infections, understand how patients become infected and how the infection evolves over time as well as identify prognostic markers for better evidence-based decisions on patient care.
- Full Text
- View/download PDF
157. SYSTEMS BASED APPROACH TO UNDERSTANDING THE ROLE OF STAPHYLOCOCCUS AUREUS IN CRS PATHOPHYSIOLOGY AND IDENTIFYING NOVEL THERAPIES.
- Author
-
Bahakim, Abdullah, Valera Cardoso, Fabian, Barbeau, Jean, Kilty, Shaun, Schwartz, Joseph, Brochiero, Emmanuelle, Rousseau, Simon, Madrenas, Joaquin, Endam, Leandra Mfuna, and Desrosiers, Martin
- Published
- 2018
158. S. AUREUS IMPAIRS LAMELLIPODIA AND CYTOSKELETAL ORGANIZATION IN NASAL EPITHELIAL CELLS.
- Author
-
Valera, Fabiana, Ruffin, Manon, Adam, Damien, Maillé, Émilie, Ibrahim, Badr, Berube, Julie, Rousseau, Simon, Brochiero, Emmanuelle, and Desrosiers, Martin
- Published
- 2018
159. EPITHELIAL REPAIR IS IMPAIRED IN CRSwNP AND IS MORE SENSITIVE TO S. AUREUS EXOPRODUCT-INDUCED DYSFUNCTIONAL REPAIR.
- Author
-
Valera, Fabiana, Ruffin, Manon, Adam, Damien, Maillé, Émilie, Ibrahim, Badr, Berube, Julie, Rousseau, Simon, Brochiero, Emmanuelle, and Desrosiers, Martin
- Published
- 2018
160. Molecular Signatures as a New Classification Scheme for CRS.
- Author
-
Al‐Mot, Sawsan, Filali‐Mouhim, Ali, Rousseau, Simon, Tardif, Valerie, Berthiaume, Yves, Desrosiers, Martin Y., and Mfuna‐Endam, Leandra
- Abstract
Objective: We have identified 2 distinct gene expression patterns (CRS1 and CRS2) in cultured epithelial cells obtained from surgical biopsies in CRS patients with and without nasal polyps (NP), which may better classify this disorder. We wished to compare the cellular infiltrate in CRS when assessed by molecular and clinical phenotypes. Method: Surgical biopsies taken from 20 CRS patients and 10 controls were divided according to their molecular signature as determined from epithelial cell cultures. Immunohistochemistry (IHC) for neutrophil elastase (NE) and major basic protein (MBP) was used to identify neutrophils and eosinophils in the epithelial and submucosal areas. Results: Groups were analyzed statistically using ANOVA and t test to identify significant differences between groups. When biopsies were assessed as function of the clinical phenotypes of CRSwNP compared with CRSsNP, while there were equal increases in the number of eosinophils and neutrophils cells in CRS patients compared with the control subjects, there was no difference between the win and sNP groups. However, when assessed according to molecular signature, while there was an increase in eosinophils in both groups, a significant increase in the number of neutrophils was only seen in the CRS2 group, suggesting a different pathogenic mechanism. Conclusion: This novel expression signature identifies a molecular phenotype of CRS that is characterized by a marked neutrophil infiltration. Assessments of disease mechanism and response to therapy according to molecular, as opposed to clinical, phenotype may identify mechanistic differences which "personalize" our management of individuals with this disease. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
161. TH17 cytokines induce human airway smooth muscle cell migration.
- Author
-
Chang, Ying, Al-Alwan, Laila, Risse, Paul-André, Roussel, Lucie, Rousseau, Simon, Halayko, Andrew J., Martin, James G., Hamid, Qutayba, and Eidelman, David H.
- Subjects
CYTOKINES ,SMOOTH muscle ,CELL migration ,ASTHMA ,PHOSPHOINOSITIDES ,MITOGEN-activated protein kinases ,NF-kappa B ,B cells - Abstract
Background: Migration of airway smooth muscle cells (ASMCs) might contribute to increased airway smooth muscle mass in asthma. T
H 17 cells and TH 17-associated cytokines are involved in the pathogenesis of asthma and might also contribute to airway remodeling. Objective: We sought to explore the possibility that migration of ASMCs might contribute to airway remodeling through the action of TH 17-related cytokines. Methods: The effect of exogenous TH 17 cytokines on ex vivo human ASMC migration was investigated by using a chemotaxis assay. The involvement of signaling pathways, including p38 mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase 1/2 MAPK, nuclear factor κB, and phosphoinositide 3-kinase, was also examined. Results: We demonstrated that IL-17A, IL-17F, and IL-22 promote migration in a dose-dependent manner. We further demonstrated that ASMCs express receptors for IL-17RA, IL-17RC, and IL-22R1. Using mAbs directed against these receptors, we confirmed that TH 17-associated cytokine–induced migration was dependent on selective receptor activation. Moreover, IL-17A and IL-17F exert their effects through signaling pathways that are distinct from those used by IL-22. The p38 MAPK inhibitor BIRB0796 inhibited the migration induced by IL-17A and IL-17F. PS1145, an inhibitor of nuclear factor κB, abolished the IL-22–induced migration. Conclusion: These data raise the possibility that TH 17-associated cytokines promote human ASMC migration in vivo and suggest an important new mechanism for the promotion of airway remodeling in asthma. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
162. A Model of Intracellular Persistence of Pseudomonas aeruginosa in Airway Epithelial Cells.
- Author
-
Malet, Julien K., Hennemann, Lisa C., Hua, Elizabeth M.-L., Faure, Emmanuel, Waters, Valerie, Rousseau, Simon, and Nguyen, Dao
- Subjects
- *
EPITHELIAL cells , *PSEUDOMONAS aeruginosa , *CYSTIC fibrosis , *CONFOCAL microscopy , *FLOW cytometry , *GENETIC disorders - Abstract
Pseudomonas aeruginosa (P.a.) is a major human pathogen capable of causing chronic infections in hosts with weakened barrier functions and host defenses, most notably airway infections commonly observed in individuals with the genetic disorder cystic fibrosis (CF). While mainly described as an extracellular pathogen, previous in vitro studies have described the molecular events leading to P.a. internalization in diverse epithelial cell types. However, the long-term fate of intracellular P.a. remains largely unknown. Here, we developed a model allowing for a better understanding of long-term (up to 120 h) intracellular bacterial survival in the airway epithelial cell line BEAS-2B. Using a tobramycin protection assay, we characterized the internalization, long-term intracellular survival, and cytotoxicity of the lab strain PAO1, as well as clinical CF isolates, and conducted analyses at the single-cell level using confocal microscopy and flow cytometry techniques. We observed that infection at low multiplicity of infection allows for intracellular survival up to 120 h post-infection without causing significant host cytotoxicity. Finally, infection with clinical isolates revealed significant strain-to-strain heterogeneity in intracellular survival, including a high persistence phenotype associated with bacterial replication within host cells. Future studies using this model will further elucidate the host and bacterial mechanisms that promote P. aeruginosa intracellular persistence in airway epithelial cells, a potentially unrecognized bacterial reservoir during chronic infections. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
163. Mass spectrometry imaging in zebrafish larvae for assessing drug safety and metabolism.
- Author
-
Asslan, Mariana, Lauzon, Nidia, Beus, Maja, Maysinger, Dusica, and Rousseau, Simon
- Subjects
- *
MATRIX-assisted laser desorption-ionization , *MEDICATION safety , *MASS spectrometry , *DRUG metabolism , *LIQUID chromatography-mass spectrometry , *BRACHYDANIO - Abstract
Drug safety assessment in the early phases of drug discovery is critical to facilitate the rapid development of novel therapeutics. Recently, teleost zebrafish (Danio rerio) has emerged as a promising vertebrate model for the assessment of drug safety. Zebrafish is a convenient model because of its small size, high fecundity, embryo transparency, and ex utero development. In this study, we developed a matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) method applied to zebrafish larvae to investigate safety and metabolism of sahaquine (Sq), an anticancer agent inhibiting histone deacetylase 6. This technique improves on prior studies using liquid chromatography-mass spectrometry (LC-MS) by adding analysis of the drug spatial distribution. Using this method, it was determined that Sq dissolved in fish water (1–2000 μM) did not reach the larval body and was mainly distributed throughout the yolk. High Sq concentration (800 μM) administered intravenously allowed the compound to reach the larval body but did not induce phenotypic abnormalities. Sq was metabolized into its glucuronidated form within 24 h and was excreted within 72 h. MALDI MSI was instrumental in showing that Sq-glucuronide was mainly formed in the gut and slightly in yolk syncytial layer, and provided valuable insights into xenobiotics elimination in zebrafish larvae. This study indicates that Sq has a good safety profile and merits further investigations in other disease models. In addition, the optimized MALDI MSI protocol provided here can be widely applied to study distribution and metabolic fate of other structurally related molecules. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
164. LasR-deficient Pseudomonas aeruginosa variants increase airway epithelial mICAM-1 expression and enhance neutrophilic lung inflammation.
- Author
-
Hennemann, Lisa C., LaFayette, Shantelle L., Malet, Julien K., Bortolotti, Perrine, Yang, Tianxiao, McKay, Geoffrey A., Houle, Daniel, Radzioch, Danuta, Rousseau, Simon, and Nguyen, Dao
- Subjects
- *
PSEUDOMONAS aeruginosa infections , *BURKHOLDERIA infections , *PNEUMONIA , *PSEUDOMONAS aeruginosa , *AIRWAY (Anatomy) , *QUORUM sensing , *CYSTIC fibrosis , *EPITHELIAL cells - Abstract
Pseudomonas aeruginosa causes chronic airway infections, a major determinant of lung inflammation and damage in cystic fibrosis (CF). Loss-of-function lasR mutants commonly arise during chronic CF infections, are associated with accelerated lung function decline in CF patients and induce exaggerated neutrophilic inflammation in model systems. In this study, we investigated how lasR mutants modulate airway epithelial membrane bound ICAM-1 (mICAM-1), a surface adhesion molecule, and determined its impact on neutrophilic inflammation in vitro and in vivo. We demonstrated that LasR-deficient strains induce increased mICAM-1 levels in airway epithelial cells compared to wild-type strains, an effect attributable to the loss of mICAM-1 degradation by LasR-regulated proteases and associated with enhanced neutrophil adhesion. In a subacute airway infection model, we also observed that lasR mutant-infected mice displayed greater airway epithelial ICAM-1 expression and increased neutrophilic pulmonary inflammation. Our findings provide new insights into the intricate interplay between lasR mutants, LasR-regulated proteases and airway epithelial ICAM-1 expression, and reveal a new mechanism involved in the exaggerated inflammatory response induced by lasR mutants. Author summary: Cystic fibrosis (CF) patients develop progressive lung disease characterized by chronic airway infections, commonly caused by the opportunistic pathogen Pseudomonas aeruginosa, and excessive non-resolving neutrophilic inflammation. Loss of function mutations of the lasR quorum sensing transcription regulator gene commonly arise during chronic P. aeruginosa infections and are associated with increased lung inflammation. In this study, we demonstrated that loss-of-function lasR mutants induced increased mICAM-1 levels on airway epithelial cells compared to wild-type P. aeruginosa strains in cell culture and in murine infection models. This effect was caused by the loss of ICAM-1 degradation by LasR-regulated secreted protease, facilitated neutrophil adhesion, and was associated with increased neutrophilic lung inflammation. Our study provides novel insights into the P. aeruginosa—airway epithelial–neutrophil interactions, and demonstrates how a common pathoadaptation of P. aeruginosa may drive lung disease progression by exacerbating inflammation. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
165. A network-informed analysis of SARS-CoV-2 and hemophagocytic lymphohistiocytosis genes' interactions points to Neutrophil extracellular traps as mediators of thrombosis in COVID-19.
- Author
-
Ding, Jun, Hostallero, David Earl, El Khili, Mohamed Reda, Fonseca, Gregory Joseph, Milette, Simon, Noorah, Nuzha, Guay-Belzile, Myriam, Spicer, Jonathan, Daneshtalab, Noriko, Sirois, Martin, Tremblay, Karine, Emad, Amin, and Rousseau, Simon
- Subjects
- *
COVID-19 , *SARS-CoV-2 , *HEMOPHAGOCYTIC lymphohistiocytosis , *GENES , *COVID-19 pandemic , *NEUTROPHILS , *GENE regulatory networks - Abstract
Abnormal coagulation and an increased risk of thrombosis are features of severe COVID-19, with parallels proposed with hemophagocytic lymphohistiocytosis (HLH), a life-threating condition associated with hyperinflammation. The presence of HLH was described in severely ill patients during the H1N1 influenza epidemic, presenting with pulmonary vascular thrombosis. We tested the hypothesis that genes causing primary HLH regulate pathways linking pulmonary thromboembolism to the presence of SARS-CoV-2 using novel network-informed computational algorithms. This approach led to the identification of Neutrophils Extracellular Traps (NETs) as plausible mediators of vascular thrombosis in severe COVID-19 in children and adults. Taken together, the network-informed analysis led us to propose the following model: the release of NETs in response to inflammatory signals acting in concert with SARS-CoV-2 damage the endothelium and direct platelet-activation promoting abnormal coagulation leading to serious complications of COVID-19. The underlying hypothesis is that genetic and/or environmental conditions that favor the release of NETs may predispose individuals to thrombotic complications of COVID-19 due to an increase risk of abnormal coagulation. This would be a common pathogenic mechanism in conditions including autoimmune/infectious diseases, hematologic and metabolic disorders. Author summary: In this manuscript we mined available genetic information on a life-threatening disease associated with hyperinflammation, hemophagocytic lymphohistiocytosis (HLH), that shares similarities with features of severe COVID-19 to identify potential molecular mechanisms of pathogenesis. This was achieved using advanced computational algorithms that generated a network of interactions linking the host gene products to the viral proteins of SARS-CoV-2, responsible for COVID-19. The information contained within the interaction network was then exploited by a different set of computer algorithms to 1) identify an inflammatory process, the release of extracellular traps from neutrophils, circulating immune cells involve in host defence, as a potential mechanism increasing the risk of abnormal coagulation in severe COVID-19 and 2) predict potential underlying conditions or co-morbidities that increases the risk of severe COVID-19. Therefore, we hypothesize that inflammatory conditions that predispose neutrophils to release extracellular traps may enhanced the likelihood of developing coagulation problems that have may have serious consequences for COVID-19 clinical trajectory. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
166. Neutrophil Extracellular Trap-Associated CEACAM1 as a Putative Therapeutic Target to Prevent Metastatic Progression of Colon Carcinoma.
- Author
-
Rayes, Roni F., Vourtzoumis, Phil, Rjeily, Marianne Bou, Seth, Rashmi, Bourdeau, France, Giannias, Betty, Berube, Julie, Yu-Hwa Huang, Rousseau, Simon, Camilleri-Broet, Sophie, Blumberg, Richard S., Beauchemin, Nicole, Najmeh, Sara, Cools-Lartigue, Jonathan, Spicer, Jonathan D., and Ferri, Lorenzo E.
- Subjects
- *
CELL adhesion molecules , *CANCER invasiveness , *COLON (Anatomy) , *NEUTROPHILS , *METASTASIS - Abstract
Neutrophils promote tumor growth and metastasis at multiple stages of cancer progression. One mechanism through which this occurs is via release of neutrophil extracellular traps (NETs). We have previously shown that NETs trap tumor cells in both the liver and the lung, increasing their adhesion and metastasis following postoperative complications. Multiple studies have since shown that NETs play a role in tumor progression and metastasis. NETs are composed of nuclear DNA-derived web-like structures decorated with neutrophil-derived proteins. However, it is unknown which, if any, of these NET-affiliated proteins is responsible for inducing the metastatic phenotype. In this study, we identify the NET-associated carcinoembryonic Ag cell adhesion molecule 1 (CEACAM1) as an essential element for this interaction. Indeed, blocking CEACAM1 on NETs, or knocking it out in a murine model, leads to a significant decrease in colon carcinoma cell adhesion, migration and metastasis. Thus, this work identifies NET-associated CEACAM1 as a putative therapeutic target to prevent the metastatic progression of colon carcinoma. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
167. The NF-κB family member RelB regulates microRNA miR-146a to suppress cigarette smoke-induced COX-2 protein expression in lung fibroblasts.
- Author
-
Zago, Michela, Rico de Souza, Angela, Hecht, Emelia, Rousseau, Simon, Hamid, Qutayba, Eidelman, David H., and Baglole, Carolyn J.
- Subjects
- *
NF-kappa B , *MICRORNA , *CIGARETTE smoke , *CYCLOOXYGENASE 2 , *GENE expression , *INFLAMMATION , *FIBROBLASTS , *TUMOR necrosis factors - Abstract
Highlights: [•] RelB suppresses cigarette smoke-induced COX-2 protein expression. [•] RelB controls the expression of miR-146a. [•] miR-146a suppresses cigarette smoke-induced COX-2 expression. [•] RelB regulation of miR-146a may protect against inflammation from cigarette smoke. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
168. The Level of p38α Mitogen-Activated Protein Kinase Activation in Airway Epithelial Cells Determines the Onset of Innate Immune Responses to Planktonic and Biofilm Pseudomonas aeruginosa.
- Author
-
Beaudoin, Trevor, LaFayette, Shantelle, Roussel, Lucie, Bérubé, Julie, Desrosiers, Martin, Nguyen, Dao, and Rousseau, Simon
- Subjects
- *
PSEUDOMONAS aeruginosa infections , *BIOFILMS , *PLANKTON , *MITOGEN-activated protein kinases , *AIRWAY (Anatomy) , *EPITHELIAL cells , *IMMUNE response , *HOSTS (Biology) - Abstract
Biofilm microcolonies of Pseudomonas aeruginosa chronically infect the airways of patients with cystic fibrosis and fuel ongoing destructive inflammation, yet the impact of the switch from planktonic to biofilm growth on host responses is poorly understood. We report that in airway epithelial cells a threshold of p38α mitogen-activated protein kinase (MAPK) activation was required to trigger neutrophil recruitment, which is influenced by extrinsic and intrinsic factors. Planktonic P. aeruginosa diffusible material (PsaDM) induced stronger p38α MAPK activation as compared to biofilm PsaDM. Biofilm PsaDM activated p38α MAPK in a Toll-like receptor–independent fashion via the lasI/lasR quorum-sensing system, but this activation was insufficient to recruit neutrophils. However, in airway epithelial cells from patients with cystic fibrosis with hypersensitivity to injurious stimuli, biofilm PsaDM activated p38α MAPK strongly enough to recruit neutrophils, which can contribute to lung injury. [ABSTRACT FROM PUBLISHER]
- Published
- 2013
- Full Text
- View/download PDF
169. Mucoid Pseudomonas aeruginosa caused by mucAmutations result in activation of TLR2 in addition to TLR5 in airway epithelial cells
- Author
-
Beaudoin, Trevor, LaFayette, Shantelle, Nguyen, Dao, and Rousseau, Simon
- Subjects
- *
PSEUDOMONAS aeruginosa , *GENETIC mutation , *TOLL-like receptors , *EPITHELIAL cells , *PHENOTYPES , *CYSTIC fibrosis , *LUNG diseases , *MITOGEN-activated protein kinases - Abstract
Abstract: The presence of the mucoid phenotype of Pseudomonas aeruginosa is a marker of poor survival in cystic fibrosis. As CF lung disease results from chronic infection leading to airway inflammation, we determined whether the switch to a mucoid phenotype by P. aeruginosa has an impact on the inflammatory response of airway epithelial cells. Exposure of airway epithelial cells to non-mucoid and mucoid P. aeruginosa-derived material leads to p38α MAPK activation, a key protein kinase involved in transmitting inflammatory signals. However, while the non-mucoid strain PAO1 activates p38α MAPK pathway solely via TLR5, the mucoid strain PACF508 activates p38α MAPK via both TLR5 and TLR2. Inactivation of mucA (the gene responsible for the mucoid phenotype) in PAO1 leads to p38α MAPK activation by both TLR2 and TLR5, as observed in the clinical mucoid isolate PACF508. Therefore, the switch to mucoid phenotype may contribute to more inflammation via TLR2 activation in addition to TLR5. Our findings highlight an important and under recognized role for TLR2 in the response of airway epithelial cells to infection. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
170. Loss of Cystic Fibrosis Transmembrane Conductance Regulator Function Enhances Activation of p38 and ERK MAPKs, Increasing Interleukin-6 Synthesis in Airway Epithelial Cells Exposed to Pseudomonas aeruginosa.
- Author
-
Bérubé, Julie, Roussel, Lucie, Nattagh, Leila, and Rousseau, Simon
- Subjects
- *
CYSTIC fibrosis , *EPITHELIAL cells , *INTERLEUKIN-6 , *PSEUDOMONAS aeruginosa , *AIRWAY (Anatomy) , *GENETIC mutation , *REACTIVE oxygen species , *DISEASES - Abstract
In cystic fibrosis (CF), the absence of functional cystic fibrosis transmembrane conductance regulator (CFTR) translates into chronic bacterial infection, excessive inflammation, tissue damage, impaired lung function and eventual death. Understanding the mechanisms underlying this vicious circle of inflammation is important to design better therapies for CF. We found in CF lung biopsies increased immunoreactivity for p38 MAPK activity markers. Moreover, when compared with their non-CF counterpart, airway epithelial cells expressing the most common mutation in CF (CFTRΔF508) were more potent at inducing neutrophil chemotaxis through increased interleukin (IL)-6 synthesis when challenged with Pseudomonas aeruginosa diffusible material. We then discovered that in CFTRΔF508 cells, the p38 and ERK MAPKs are hyperactivated in response to P. aeruginosa diffusible material, leading to increased IL-6 mRNA expression and stability. Moreover, although TLR5 contributes to p38 MAPK activation upon P. aeruginosa challenge, it only played a weak role in IL-6 synthesis. Instead, we found that the production of reactive oxygen species is essential for IL-6 synthesis in response to P. aeruginosa diffusible material. Finally, we uncovered that in CFTRΔF508 cells, the extracellular glutathione levels are decreased, leading to a greater sensitivity to reactive oxygen species, providing an explanation for the hyperactivation of the p38 and ERK MAPKs and increased IL-6 synthesis. Taken together, our study has characterized a mechanism whereby the CFTRΔF508 mutation in airway epithelial cells contributes to increase inflammation of the airways. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
171. Circulating IL-17F, but not IL-17A, is elevated in severe COVID-19 and leads to an ERK1/2 and p38 MAPK-dependent increase in ICAM-1 cell surface expression and neutrophil adhesion on endothelial cells.
- Author
-
Bédard-Matteau J, Soulé A, Liu KY, Fourcade L, Fraser DD, Emad A, and Rousseau S
- Subjects
- Humans, Male, Female, Middle Aged, SARS-CoV-2 immunology, SARS-CoV-2 physiology, MAP Kinase Signaling System immunology, Aged, Adult, Prospective Studies, Severity of Illness Index, Interleukin-17 blood, Interleukin-17 metabolism, Neutrophils immunology, Neutrophils metabolism, COVID-19 immunology, COVID-19 blood, Intercellular Adhesion Molecule-1 blood, Intercellular Adhesion Molecule-1 metabolism, p38 Mitogen-Activated Protein Kinases metabolism, Cell Adhesion, Endothelial Cells metabolism, Endothelial Cells immunology
- Abstract
Background: Severe COVID-19 is associated with neutrophilic inflammation and immunothrombosis. Several members of the IL-17 cytokine family have been associated with neutrophilic inflammation and activation of the endothelium. Therefore, we investigated whether these cytokines were associated with COVID-19., Methods: We investigated the association between COVID-19 and circulating plasma levels of IL-17 cytokine family members in participants to the Biobanque québécoise de la COVID-19 (BQC19), a prospective observational cohort and an independent cohort from Western University (London, Ontario). We measured the in vitro impact of IL-17F on intercellular adhesion molecule 1 (ICAM-1) cell surface expression and neutrophil adhesion on endothelial cells in culture. The contribution of two Mitogen Activated Protein Kinase (MAPK) pathways was determined using small molecule inhibitors PD184352 (a MKK1/MKK2 inhibitor) and BIRB0796 (a p38 MAPK inhibitor)., Results: We found increased IL-17D and IL-17F plasma levels when comparing SARS-CoV-2-positive vs negative hospitalized participants. Moreover, increased plasma levels of IL-17D, IL-17E and IL-17F were noted when comparing severe versus mild COVID-19. IL-17F, but not IL-17A, was significantly elevated in people with COVID-19 compared to healthy controls and with more severe disease. In vitro work on endothelial cells treated with IL-17F for 24h showed an increase cell surface expression of ICAM-1 accompanied by neutrophil adhesion. The introduction of two MAPK inhibitors significantly reduced the binding of neutrophils while also reducing ICAM-1 expression at the surface level of endothelial cells, but not its intracellular expression., Discussion: Overall, these results have identified an association between two cytokines of the IL-17 family (IL-17D and IL-17F) with COVID-19 and disease severity. Considering that IL-17F stimulation promotes neutrophil adhesion to the endothelium in a MAPK-dependent manner, it is attractive to speculate that this pathway may contribute to pathogenic immunothrombosis in concert with other molecular effectors., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Bédard-Matteau, Soulé, Liu, Fourcade, Fraser, Emad and Rousseau.)
- Published
- 2024
- Full Text
- View/download PDF
172. Intracellular Pseudomonas aeruginosa within the Airway Epithelium of Cystic Fibrosis Lung Tissues.
- Author
-
Malet K, Faure E, Adam D, Donner J, Liu L, Pilon SJ, Fraser R, Jorth P, Newman DK, Brochiero E, Rousseau S, and Nguyen D
- Subjects
- Humans, Female, Male, Adult, Young Adult, Epithelial Cells microbiology, Cystic Fibrosis microbiology, Cystic Fibrosis complications, Pseudomonas aeruginosa, Respiratory Mucosa microbiology, Respiratory Mucosa pathology, Lung Transplantation, Pseudomonas Infections microbiology, Lung microbiology, Lung pathology
- Abstract
Rationale: Pseudomonas aeruginosa is the major bacterial pathogen colonizing the airways of adult patients with cystic fibrosis (CF) and causes chronic infections that persist despite antibiotic therapy. Intracellular bacteria may represent an unrecognized reservoir of bacteria that evade the immune system and antibiotic therapy. Although the ability of P. aeruginosa to invade and survive within epithelial cells has been described in vitro in different epithelial cell models, evidence of this intracellular lifestyle in human lung tissues is currently lacking. Objectives: To detect and characterize intracellular P. aeruginosa in CF airway epithelium from human lung explant tissues. Methods: We sampled lung explant tissues from patients with CF undergoing lung transplantation and non-CF lung donor control tissue. We analyzed lung tissue sections for the presence of intracellular P. aeruginosa using quantitative culture and microscopy, in parallel to histopathology and airway morphometry. Measurements and Main Results: P. aeruginosa was isolated from the lungs of seven patients with CF undergoing lung transplantation. Microscopic assessment revealed the presence of intracellular P. aeruginosa within airway epithelial cells in three of the seven patients analyzed at a varying but low frequency. We observed those events occurring in lung regions with high bacterial burden. Conclusions: This is the first study describing the presence of intracellular P. aeruginosa in CF lung tissues. Although intracellular P. aeruginosa in airway epithelial cells is likely relatively rare, our findings highlight the plausible occurrence of this intracellular bacterial reservoir in chronic CF infections.
- Published
- 2024
- Full Text
- View/download PDF
173. Plasma taurine level is linked to symptom burden and clinical outcomes in post-COVID condition.
- Author
-
Khoramjoo M, Wang K, Srinivasan K, Gheblawi M, Mandal R, Rousseau S, Wishart D, Prasad V, Richer L, Cheung AM, and Oudit GY
- Subjects
- Humans, Female, Male, Middle Aged, Adult, Biomarkers blood, Aged, Post-Acute COVID-19 Syndrome, Case-Control Studies, Metabolome, Symptom Burden, Taurine blood, COVID-19 blood, COVID-19 complications, SARS-CoV-2 isolation & purification
- Abstract
Background: A subset of individuals (10-20%) experience post-COVID condition (PCC) subsequent to initial SARS-CoV-2 infection, which lacks effective treatment. PCC carries a substantial global burden associated with negative economic and health impacts. This study aims to evaluate the association between plasma taurine levels with self-reported symptoms and adverse clinical outcomes in patients with PCC., Methods and Findings: We analyzed the plasma proteome and metabolome of 117 individuals during their acute COVID-19 hospitalization and at the convalescence phase six-month post infection. Findings were compared with 28 age and sex-matched healthy controls. Plasma taurine levels were negatively associated with PCC symptoms and correlated with markers of inflammation, tryptophan metabolism, and gut dysbiosis. Stratifying patients based on the trajectories of plasma taurine levels during six-month follow-up revealed a significant association with adverse clinical events. Increase in taurine levels during the transition to convalescence were associated with a reduction in adverse events independent of comorbidities and acute COVID-19 severity. In a multivariate analysis, increased plasma taurine level between acute and convalescence phase was associated with marked protection from adverse clinical events with a hazard ratio of 0.13 (95% CI: 0.05-0.35; p<0.001)., Conclusions: Taurine emerges as a promising predictive biomarker and potential therapeutic target in PCC. Taurine supplementation has already demonstrated clinical benefits in various diseases and warrants exploration in large-scale clinical trials for alleviating PCC., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2024 Khoramjoo et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
174. Development of a long noncoding RNA-based machine learning model to predict COVID-19 in-hospital mortality.
- Author
-
Devaux Y, Zhang L, Lumley AI, Karaduzovic-Hadziabdic K, Mooser V, Rousseau S, Shoaib M, Satagopam V, Adilovic M, Srivastava PK, Emanueli C, Martelli F, Greco S, Badimon L, Padro T, Lustrek M, Scholz M, Rosolowski M, Jordan M, Brandenburger T, Benczik B, Agg B, Ferdinandy P, Vehreschild JJ, Lorenz-Depiereux B, Dörr M, Witzke O, Sanchez G, Kul S, Baker AH, Fagherazzi G, Ollert M, Wereski R, Mills NL, and Firat H
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Europe epidemiology, Canada epidemiology, Cohort Studies, Aged, 80 and over, Adult, COVID-19 mortality, COVID-19 virology, COVID-19 genetics, Machine Learning, RNA, Long Noncoding genetics, Hospital Mortality, SARS-CoV-2 genetics, SARS-CoV-2 isolation & purification
- Abstract
Tools for predicting COVID-19 outcomes enable personalized healthcare, potentially easing the disease burden. This collaborative study by 15 institutions across Europe aimed to develop a machine learning model for predicting the risk of in-hospital mortality post-SARS-CoV-2 infection. Blood samples and clinical data from 1286 COVID-19 patients collected from 2020 to 2023 across four cohorts in Europe and Canada were analyzed, with 2906 long non-coding RNAs profiled using targeted sequencing. From a discovery cohort combining three European cohorts and 804 patients, age and the long non-coding RNA LEF1-AS1 were identified as predictive features, yielding an AUC of 0.83 (95% CI 0.82-0.84) and a balanced accuracy of 0.78 (95% CI 0.77-0.79) with a feedforward neural network classifier. Validation in an independent Canadian cohort of 482 patients showed consistent performance. Cox regression analysis indicated that higher levels of LEF1-AS1 correlated with reduced mortality risk (age-adjusted hazard ratio 0.54, 95% CI 0.40-0.74). Quantitative PCR validated LEF1-AS1's adaptability to be measured in hospital settings. Here, we demonstrate a promising predictive model for enhancing COVID-19 patient management., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
175. Sex differences in vascular endothelial function related to acute and long COVID-19.
- Author
-
Kitselman A K, Bédard-Matteau J, Rousseau S, Tabrizchi R, and Daneshtalab N
- Subjects
- Humans, Female, Male, Post-Acute COVID-19 Syndrome, SARS-CoV-2, Sex Characteristics, COVID-19, Fatigue Syndrome, Chronic epidemiology, Vascular Diseases
- Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus has been at the forefront of health sciences research since its emergence in China in 2019 that quickly led to a global pandemic. As a result of this research, and the large numbers of infected patients globally, there were rapid enhancements made in our understanding of Coronavirus disease 2019 (COVID-19) pathology, including its role in the development of uncontrolled immune responses and its link to the development of endotheliitis and endothelial dysfunction. There were also some noted differences in the rate and severity of infection between males and females with acute COVID. Some individuals infected with SARS-CoV-2 also experience long-COVID, an important hallmark symptom of this being Myalgic Encephalomyelitis-Chronic Fatigue Syndrome (ME-CFS), also experienced differently between males and females. The purpose of this review is to discuss the impact of sex on the vasculature during acute and long COVID-19, present any link between ME-CFS and endothelial dysfunction, and provide evidence for the relationship between ME-CFS and the immune system. We also will delineate biological sex differences observed in other post viral infections and, assess if sex differences exist in how the immune system responds to viral infection causing ME-CFS., Competing Interests: Declaration of Competing Interest None to declare., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
176. The Effect of Corticosteroids on Sinus Microbiota in Chronic Rhinosinusitis Patients with Nasal Polyposis.
- Author
-
Alammar Y, Rousseau S, Desrosiers M, and Tewfik MA
- Subjects
- Humans, Case-Control Studies, Pilot Projects, Adrenal Cortex Hormones therapeutic use, Chronic Disease, Anti-Bacterial Agents therapeutic use, Rhinitis complications, Sinusitis complications, Nasal Polyps drug therapy, Nasal Polyps complications, Microbiota
- Abstract
Background: Chronic rhinosinusitis with nasal polyposis (CRSwNP) is a multifactorial disease with no known single cause, but it is thought that bacteria play a role in the disease process., Objective: This pilot study aims to assess the longitudinal effect of corticosteroid therapy on sinus microbiota in chronic rhinosinusitis patients with nasal polyposis (CRSwNP)., Methods: A longitudinal prospective case-control study was done on patients with CRSwNP and healthy controls. Patients with CRSwNP were randomly allocated to a corticosteroids and antibiotics treatment group (CRSwNP-SA) or a corticosteroid-only treatment group (CRSwNP-S). Data were collected at three-time points (before treatment, 1, and 3 months after treatment). Specimens were cultured and matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry (MS) was used as a bacterial detection method., Results: Data from 29 patients with CRSwNP (16 CRSwNP-SA and 13 CRSwNP-S) was compared to 15 healthy subjects. Patients reported significant symptom improvement initially (1 month), but not in the long-term (3 months). This result was found in both treatment groups, whether or not antibiotics were used. After 3 months from treatment, the prevalence of Corynebacterium genera tended to increase in the CRSwNP-SA, while Staphylococcus and Gram-negative genera (Pseudomonas) tended to increase in the CRSwNP-S. Smoking, aspirin sensitivity, and previous endoscopic sinus surgery were found to be co-factors significantly associated with the response to systemic corticosteroid therapy., Conclusion: In this pilot study, both treatment options were effective to improve symptoms in the short-term but not in the long-term, and were not linked to any clear sinus microbiota response. As a result, this study supports the avoidance of systemic antibiotics without evidence of active infection., Competing Interests: Declaration of Conflicting InterestsThe author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
- Published
- 2023
- Full Text
- View/download PDF
177. Validation of ANG-1 and P-SEL as biomarkers of post-COVID-19 conditions using data from the Biobanque québécoise de la COVID-19 (BQC-19).
- Author
-
Yamga E, Soulé A, Piché A, Emad A, Durand M, and Rousseau S
- Abstract
The quest for understanding and managing the long-term effects of COVID-19, often referred to as Long COVID or post-COVID-19 condition (PCC), remains an active research area. Recent findings highlighted angiopoietin-1 (ANG-1) and p-selectin (P-SEL) as potential diagnostic markers, but validation is essential, given the inconsistency in COVID-19 biomarker studies. Leveraging the biobanque québécoise de la COVID-19 (BQC19) biobank, we analyzed the data of 249 participants. Both ANG-1 and P-SEL levels were significantly higher in patients with PCC participants compared with control subjects at 3 months using the Mann-Whitney U test. We managed to reproduce and validate the findings, emphasizing the importance of collaborative biobanking efforts in enhancing the reproducibility and credibility of Long COVID research outcomes., (© 2023. BioMed Central Ltd., part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
178. Correction: TPL2-mediated activation of ERK1 and ERK2 regulates the processing of pre-TNFα in LPS-stimulated macrophages.
- Author
-
Rousseau S, Papoutsopoulou M, Symons A, Cook D, Lucocq JM, Prescott AR, O'Garra A, Ley SC, and Cohen P
- Published
- 2022
- Full Text
- View/download PDF
179. Inhibition of LPS-mediated TLR4 activation abrogates gastric adenocarcinoma-associated peritoneal metastasis.
- Author
-
Sangwan V, Al-Marzouki L, Pal S, Stavrakos V, Alzahrani M, Antonatos D, Nevo Y, Camilleri-Broët S, Rayes R, Bourdeau F, Giannias B, Bertos N, Bailey S, Rousseau S, Cools-Lartigue J, Spicer JD, and Ferri L
- Subjects
- Animals, Escherichia coli metabolism, Humans, Lipopolysaccharides pharmacology, Peritoneum, Toll-Like Receptor 4 metabolism, Adenocarcinoma, Peritoneal Neoplasms, Stomach Neoplasms
- Abstract
Surgical resection, the cornerstone of curative intent treatment for gastric adenocarcinoma, is associated with a high rate of infection-related post-operative complications, leading to an increased incidence of metastasis to the peritoneum. However, the mechanisms underlying this process are poorly understood. Lipopolysaccharide (LPS), an antigen from Gram-negative bacteria, represents a potential mechanism via induction of local and systemic inflammation through activation of Toll-like receptor (TLR). Here, we use both a novel ex vivo model of peritoneal metastasis and in vivo animal models to assess gastric cancer cell adhesion to peritoneum both before and after inhibition of the TLR4 pathway. We demonstrate that activation of TLR4 by either LPS or Gram-negative bacteria (E. coli) significantly increases the adherence of gastric cancer cells to human peritoneal mesothelial cells, and that this increased adherence is abrogated by inhibition of the TLR4 signal cascade and downstream TAK1 and MEK1/2 pathways. We also demonstrate that the influence of LPS on adherence extends to peritoneal tissue and metastatic spread. Furthermore, we show that loss of TLR4 at the site of metastasis reduces tumor cell adhesion, implicating the TLR4 signaling cascade in potentiating metastatic adhesion and peritoneal spread. These results identify potential therapeutic targets for the clinical management of patients undergoing resection for gastric cancer., (© 2021. The Author(s), under exclusive licence to Springer Nature B.V.)
- Published
- 2022
- Full Text
- View/download PDF
180. Second-Shell Amino Acid R266 Helps Determine N -Succinylamino Acid Racemase Reaction Specificity in Promiscuous N -Succinylamino Acid Racemase/ o -Succinylbenzoate Synthase Enzymes.
- Author
-
Truong DP, Rousseau S, Machala BW, Huddleston JP, Zhu M, Hull KG, Romo D, Raushel FM, Sacchettini JC, and Glasner ME
- Subjects
- Amino Acid Isomerases genetics, Amino Acid Sequence, Amino Acid Substitution, Amycolatopsis enzymology, Amycolatopsis genetics, Bacterial Proteins chemistry, Bacterial Proteins genetics, Bacterial Proteins metabolism, Biocatalysis, Carbon-Carbon Lyases genetics, Catalytic Domain genetics, Conserved Sequence, Crystallography, X-Ray, Enzyme Stability genetics, Evolution, Molecular, Kinetics, Models, Molecular, Mutagenesis, Site-Directed, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Substrate Specificity, Amino Acid Isomerases chemistry, Amino Acid Isomerases metabolism, Carbon-Carbon Lyases chemistry, Carbon-Carbon Lyases metabolism
- Abstract
Catalytic promiscuity is the coincidental ability to catalyze nonbiological reactions in the same active site as the native biological reaction. Several lines of evidence show that catalytic promiscuity plays a role in the evolution of new enzyme functions. Thus, studying catalytic promiscuity can help identify structural features that predispose an enzyme to evolve new functions. This study identifies a potentially preadaptive residue in a promiscuous N -succinylamino acid racemase/ o -succinylbenzoate synthase (NSAR/OSBS) enzyme from Amycolatopsis sp. T-1-60. This enzyme belongs to a branch of the OSBS family which includes many catalytically promiscuous NSAR/OSBS enzymes. R266 is conserved in all members of the NSAR/OSBS subfamily. However, the homologous position is usually hydrophobic in other OSBS subfamilies, whose enzymes lack NSAR activity. The second-shell amino acid R266 is close to the catalytic acid/base K263, but it does not contact the substrate, suggesting that R266 could affect the catalytic mechanism. Mutating R266 to glutamine in Amycolatopsis NSAR/OSBS profoundly reduces NSAR activity but moderately reduces OSBS activity. This is due to a 1000-fold decrease in the rate of proton exchange between the substrate and the general acid/base catalyst K263. This mutation is less deleterious for the OSBS reaction because K263 forms a cation-π interaction with the OSBS substrate and/or the intermediate, rather than acting as a general acid/base catalyst. Together, the data explain how R266 contributes to NSAR reaction specificity and was likely an essential preadaptation for the evolution of NSAR activity.
- Published
- 2021
- Full Text
- View/download PDF
181. Identification of transcriptional regulatory network associated with response of host epithelial cells to SARS-CoV-2.
- Author
-
Su C, Rousseau S, and Emad A
- Subjects
- A549 Cells, Cell Line, Epithelial Cells chemistry, Epithelial Cells cytology, Epithelial Cells virology, Gene Expression Regulation, Humans, Models, Biological, Systems Biology, COVID-19 genetics, Gene Expression Profiling methods, SARS-CoV-2 pathogenicity, Sequence Analysis, RNA methods
- Abstract
Identification of transcriptional regulatory mechanisms and signaling networks involved in the response of host cells to infection by SARS-CoV-2 is a powerful approach that provides a systems biology view of gene expression programs involved in COVID-19 and may enable the identification of novel therapeutic targets and strategies to mitigate the impact of this disease. In this study, our goal was to identify a transcriptional regulatory network that is associated with gene expression changes between samples infected by SARS-CoV-2 and those that are infected by other respiratory viruses to narrow the results on those enriched or specific to SARS-CoV-2. We combined a series of recently developed computational tools to identify transcriptional regulatory mechanisms involved in the response of epithelial cells to infection by SARS-CoV-2, and particularly regulatory mechanisms that are specific to this virus when compared to other viruses. In addition, using network-guided analyses, we identified kinases associated with this network. The results identified pathways associated with regulation of inflammation (MAPK14) and immunity (BTK, MBX) that may contribute to exacerbate organ damage linked with complications of COVID-19. The regulatory network identified herein reflects a combination of known hits and novel candidate pathways supporting the novel computational pipeline presented herein to quickly narrow down promising avenues of investigation when facing an emerging and novel disease such as COVID-19., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
182. Bedaquiline reprograms central metabolism to reveal glycolytic vulnerability in Mycobacterium tuberculosis.
- Author
-
Mackenzie JS, Lamprecht DA, Asmal R, Adamson JH, Borah K, Beste DJV, Lee BS, Pethe K, Rousseau S, Krieger I, Sacchettini JC, Glasgow JN, and Steyn AJC
- Subjects
- Antitubercular Agents pharmacology, Bacterial Proteins metabolism, Carbon Cycle drug effects, Citric Acid Cycle drug effects, Energy Metabolism drug effects, Glyoxylates, Mycobacterium tuberculosis genetics, Oxidative Phosphorylation, Tuberculosis microbiology, Anti-Bacterial Agents pharmacology, Diarylquinolines pharmacology, Glycolysis drug effects, Mycobacterium tuberculosis drug effects, Mycobacterium tuberculosis metabolism
- Abstract
The approval of bedaquiline (BDQ) for the treatment of tuberculosis has generated substantial interest in inhibiting energy metabolism as a therapeutic paradigm. However, it is not known precisely how BDQ triggers cell death in Mycobacterium tuberculosis (Mtb). Using
13 C isotopomer analysis, we show that BDQ-treated Mtb redirects central carbon metabolism to induce a metabolically vulnerable state susceptible to genetic disruption of glycolysis and gluconeogenesis. Metabolic flux profiles indicate that BDQ-treated Mtb is dependent on glycolysis for ATP production, operates a bifurcated TCA cycle by increasing flux through the glyoxylate shunt, and requires enzymes of the anaplerotic node and methylcitrate cycle. Targeting oxidative phosphorylation (OXPHOS) with BDQ and simultaneously inhibiting substrate level phosphorylation via genetic disruption of glycolysis leads to rapid sterilization. Our findings provide insight into the metabolic mechanism of BDQ-induced cell death and establish a paradigm for the development of combination therapies that target OXPHOS and glycolysis.- Published
- 2020
- Full Text
- View/download PDF
183. Azithromycin Downregulates Gene Expression of IL-1β and Pathways Involving TMPRSS2 and TMPRSS11D Required by SARS-CoV-2.
- Author
-
Renteria AE, Mfuna Endam L, Adam D, Filali-Mouhim A, Maniakas A, Rousseau S, Brochiero E, Gallo S, and Desrosiers M
- Subjects
- Betacoronavirus pathogenicity, COVID-19, Cells, Cultured, Chronic Disease, Coronavirus Infections immunology, Coronavirus Infections metabolism, Coronavirus Infections virology, Down-Regulation, Host-Pathogen Interactions, Humans, Interleukin-1beta genetics, Male, Membrane Proteins genetics, Nasal Mucosa immunology, Nasal Mucosa metabolism, Pandemics, Pilot Projects, Pneumonia, Viral immunology, Pneumonia, Viral metabolism, Pneumonia, Viral virology, Rhinitis drug therapy, Rhinitis immunology, Rhinitis metabolism, SARS-CoV-2, Serine Endopeptidases genetics, Serine Proteases genetics, Signal Transduction, Sinusitis drug therapy, Sinusitis immunology, Sinusitis metabolism, COVID-19 Drug Treatment, Anti-Inflammatory Agents pharmacology, Azithromycin pharmacology, Coronavirus Infections drug therapy, Interleukin-1beta metabolism, Membrane Proteins metabolism, Nasal Mucosa drug effects, Pneumonia, Viral drug therapy, Serine Endopeptidases metabolism, Serine Proteases metabolism
- Published
- 2020
- Full Text
- View/download PDF
184. Targeting potential drivers of COVID-19: Neutrophil extracellular traps.
- Author
-
Barnes BJ, Adrover JM, Baxter-Stoltzfus A, Borczuk A, Cools-Lartigue J, Crawford JM, Daßler-Plenker J, Guerci P, Huynh C, Knight JS, Loda M, Looney MR, McAllister F, Rayes R, Renaud S, Rousseau S, Salvatore S, Schwartz RE, Spicer JD, Yost CC, Weber A, Zuo Y, and Egeblad M
- Subjects
- COVID-19, Coronavirus Infections complications, Cytokines metabolism, Humans, Pandemics, Pneumonia, Viral complications, SARS-CoV-2, Betacoronavirus, Coronavirus Infections pathology, Extracellular Traps, Lung Diseases etiology, Lung Diseases metabolism, Lung Diseases pathology, Neutrophils pathology, Pneumonia, Viral pathology
- Abstract
Coronavirus disease 2019 (COVID-19) is a novel, viral-induced respiratory disease that in ∼10-15% of patients progresses to acute respiratory distress syndrome (ARDS) triggered by a cytokine storm. In this Perspective, autopsy results and literature are presented supporting the hypothesis that a little known yet powerful function of neutrophils-the ability to form neutrophil extracellular traps (NETs)-may contribute to organ damage and mortality in COVID-19. We show lung infiltration of neutrophils in an autopsy specimen from a patient who succumbed to COVID-19. We discuss prior reports linking aberrant NET formation to pulmonary diseases, thrombosis, mucous secretions in the airways, and cytokine production. If our hypothesis is correct, targeting NETs directly and/or indirectly with existing drugs may reduce the clinical severity of COVID-19., Competing Interests: Disclosures: M.R. Looney reported "other" from Neutrolis outside the submitted work. R.E. Schwartz is a Sponsored Advisory Board Member for Miromatrix Inc. J.D. Spicer reported personal fees from Bristol Myers Squibb, personal fees from Astra Zeneca, personal fees from Merck, personal fees from Trans-Hit Bio, and non-financial support from Astra Zeneca outside the submitted work. C.C. Yost reports a grant from PEEL Therapeutics, Inc. during the conduct of the study; in addition, C.C. Yost authors a US patent (patent no. 10,232,023 B2) held by the University of Utah for the use of NET-inhibitory peptides for "treatment of and prophylaxis against inflammatory disorders." PEEL Therapeutics, Inc. has exclusive licensing rights. M. Egeblad reported "other" from Santhera during the conduct of the study; and consulted for CytomX in 2019. No other disclosures were reported., (© 2020 Barnes et al.)
- Published
- 2020
- Full Text
- View/download PDF
185. Activation of the pattern recognition receptor NOD1 augments colon cancer metastasis.
- Author
-
Jiang HY, Najmeh S, Martel G, MacFadden-Murphy E, Farias R, Savage P, Leone A, Roussel L, Cools-Lartigue J, Gowing S, Berube J, Giannias B, Bourdeau F, Chan CHF, Spicer JD, McClure R, Park M, Rousseau S, and Ferri LE
- Subjects
- Adenocarcinoma pathology, Animals, Cell Adhesion, Cell Line, Cell Movement, Colonic Neoplasms pathology, Humans, Male, Mice, Mice, Inbred C57BL, Neoplasm Metastasis, p38 Mitogen-Activated Protein Kinases metabolism, Adenocarcinoma metabolism, Colonic Neoplasms metabolism, Nod1 Signaling Adaptor Protein physiology
- Abstract
While emerging data suggest nucleotide oligomerization domain receptor 1 (NOD1), a cytoplasmic pattern recognition receptor, may play an important and complementary role in the immune response to bacterial infection, its role in cancer metastasis is entirely unknown. Hence, we sought to determine the effects of NOD1 on metastasis. NOD1 expression in paired human primary colon cancer, human and murine colon cancer cells were determined using immunohistochemistry and immunoblotting (WB). Clinical significance of NOD1 was assessed using TCGA survival data. A series of in vitro and in vivo functional assays, including adhesion, migration, and metastasis, was conducted to assess the effect of NOD1. C12-iE-DAP, a highly selective NOD1 ligand derived from gram-negative bacteria, was used to activate NOD1. ML130, a specific NOD1 inhibitor, was used to block C12-iE-DAP stimulation. Stable knockdown (KD) of NOD1 in human colon cancer cells (HT29) was constructed with shRNA lentiviral transduction and the functional assays were thus repeated. Lastly, the predominant signaling pathway of NOD1-activation was identified using WB and functional assays in the presence of specific kinase inhibitors. Our data demonstrate that NOD1 is highly expressed in human colorectal cancer (CRC) and human and murine CRC cell lines. Clinically, we demonstrate that this increased NOD1 expression negatively impacts survival in patients with CRC. Subsequently, we identify NOD1 activation by C12-iE-DAP augments CRC cell adhesion, migration and metastasis. These effects are predominantly mediated via the p38 mitogen activated protein kinase (MAPK) pathway. This is the first study implicating NOD1 in cancer metastasis, and thus identifying this receptor as a putative therapeutic target.
- Published
- 2020
- Full Text
- View/download PDF
186. Gram-Negative Pneumonia Augments Non-Small Cell Lung Cancer Metastasis through Host Toll-like Receptor 4 Activation.
- Author
-
Gowing SD, Chow SC, Cools-Lartigue JJ, Chen CB, Najmeh S, Goodwin-Wilson M, Jiang HY, Bourdeau F, Beauchamp A, Angers I, Giannias B, Spicer JD, Rousseau S, Qureshi ST, and Ferri LE
- Subjects
- Animals, Bronchi pathology, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Epithelial Cells pathology, Escherichia coli isolation & purification, Escherichia coli Infections metabolism, Escherichia coli Infections microbiology, Humans, Liver Neoplasms metabolism, Liver Neoplasms microbiology, Lung Neoplasms metabolism, Lung Neoplasms pathology, Male, Mice, Mice, Inbred C57BL, Neoplasm Metastasis, Pneumonia, Bacterial metabolism, Pneumonia, Bacterial microbiology, Carcinoma, Non-Small-Cell Lung microbiology, Escherichia coli Infections pathology, Liver Neoplasms secondary, Lung Neoplasms microbiology, Pneumonia, Bacterial pathology, Toll-Like Receptor 4 metabolism
- Abstract
Introduction: Surgery is essential for cure of early-stage non-small cell lung cancer (NSCLC). Rates of postoperative bacterial pneumonias, however, remain high, and clinical data suggests that post-operative infectious complications confer an increased risk for metastasis. Toll-like receptors (TLRs) mediate the inflammatory response to infection by recognizing evolutionarily conserved bacterial structures at the surface of numerous pulmonary cell types; yet, little is known about how host TLR activation influences NSCLC metastasis. TLR4 recognizes gram-negative bacterium lipopolysaccharide activating the innate immune system., Methods: C57BL/6 and TLR4 knockout murine airways were inoculated with Escherichia coli or lipopolysaccharide. Hepatic metastasis assays and intravital microscopy were performed. Bronchoepithelial conditioned media was generated through coincubation of bronchoepithelial cells with TLR4 activating Escherichia coli or lipopolysaccharide. Subsequently, H59 NSCLC were stimulated with conditioned media and subject to various adhesion assays., Results: We demonstrate that gram-negative Escherichia coli pneumonia augments the formation of murine H59 NSCLC liver metastases in C57BL/6 mice through TLR4 activation. Additionally, infected C57BL/6 mice demonstrate increased H59 NSCLC in vivo hepatic sinusoidal adhesion compared with negative controls, a response that is significantly diminished in TLR4 knockout mice. Similarly, intratracheal injection of purified TLR4 activating lipopolysaccharide increases in vivo adhesion of H59 cells to murine hepatic sinusoids. Furthermore, H59 cells incubated with bronchoepithelial conditioned medium show increased cell adhesion to in vitro extracellular matrix proteins and in vivo hepatic sinusoids through a mechanism dependent on bronchoepithelial TLR4 activation and interleukin-6 secretion., Conclusion: TLR4 is a viable therapeutic target for NSCLC metastasis augmented by gram-negative pneumonia., (Copyright © 2019 International Association for the Study of Lung Cancer. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
187. Staphylococcus aureus impairs sinonasal epithelial repair: Effects in patients with chronic rhinosinusitis with nasal polyps and control subjects.
- Author
-
Valera FCP, Ruffin M, Adam D, Maillé É, Ibrahim B, Berube J, Rousseau S, Brochiero E, and Desrosiers MY
- Subjects
- Adult, Aged, Amides pharmacology, Cells, Cultured, Chronic Disease, Cytoskeleton metabolism, Humans, Male, Middle Aged, Paranasal Sinuses microbiology, Pyridines pharmacology, Respiratory Mucosa pathology, Wound Healing, rho-Associated Kinases metabolism, Nasal Polyps immunology, Paranasal Sinuses pathology, Respiratory Mucosa physiology, Rhinitis immunology, Sinusitis immunology, Staphylococcal Infections immunology, Staphylococcus aureus physiology
- Abstract
Background: The effect of Staphylococcus aureus on nasal epithelial repair has never been assessed in patients with chronic rhinosinusitis with nasal polyps (CRSwNP)., Objective: This study aimed to determine whether (1) nasal epithelial cell cultures from patients with CRSwNP and control subjects repair differently; (2) S aureus exoproducts compromise nasal epithelial repair; (3) S aureus alters lamellipodial dynamics; and (4) deleterious effects could be counteracted by the Rho-associated coiled-coil kinase inhibitor Y-27632., Methods: Primary nasal epithelial cells (pNECs) collected during surgeries were cultured and injured under 3 conditions: (1) basal conditions, (2) exposed to S aureus exoproducts, and (3) exposed to S aureus exoproducts and Y-27632. Epithelial repair, lamellipodial dynamics, and cytoskeletal organization were assessed., Results: Under basal conditions, pNEC cultures from patients with CRSwNP presented significantly lower repair rates and reduced lamellipodial protrusion length and velocity than those from control subjects. S aureus exoproducts significantly decreased repair rates and protrusion dynamics in both control subjects and patients with CRSwNP; however, the effect of S aureus on cell protrusions was more sustained over time in patients with CRSwNP. Under basal conditions, immunofluorescence assays showed significantly reduced percentages of cells with lamellipodia at the wound edge in patients with CRSwNP compared with control subjects. S aureus altered cell polarity and decreased the percentage of cells with lamellipodia in both groups. Finally, Y-27632 prevented the deleterious effects of S aureus exoproducts on CRSwNP repair rates, as well as on lamellipodial dynamics and formation., Conclusions: S aureus exoproducts significantly alter epithelial repair and lamellipodial dynamics on pNECs, and this impairment was more pronounced in patients with CRSwNP. Importantly, Y-27632 restored epithelial repair and lamellipodial dynamics in the presence of S aureus exoproducts., (Copyright © 2018 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
188. C-Reactive Protein in Stable Cystic Fibrosis: An Additional Indicator of Clinical Disease Activity and Risk of Future Pulmonary Exacerbations.
- Author
-
Matouk E, Nguyen D, Benedetti A, Bernier J, Gruber J, Landry J, Rousseau S, Ahlgren HG, Lands LC, Wojewodka G, and Radzioch D
- Abstract
Introduction: In stable adult cystic fibrosis (CF) patients, we assessed the role of baseline high sensitivity C-reactive protein (hs-CRP) on CF clinical variables and frequency of intravenous (IV) treated pulmonary exacerbations (PExs) 1-year post-baseline., Methods: We recruited 51 clinically stable CF patients from our Adult CF Center. We incorporated collected parameters into Matouk CF clinical score and CF questionnaire-revised quality of life score (QOL). We used the clinical minus complications subscores as a clinical disease activity score (CDAS). We dichotomized our patients according to the cohort median baseline hs-CRP of 5.2 mg/L., Results: Patients in the high hs-CRP group (≥ 5.2 mg/L) demonstrated worse CDAS (r=0.67, p=0.0001) and QOL scores (r=0.57, p=0.0017) at a given FEV
1 % predicted. In both hs-CRP groups, prior-year IV-treated PExs and baseline CDASs were significant predictors of future IV-treated PExs. Interestingly, the association between baseline CDAS and future PExs frequency was more robust in the high compared to the low hs-CRP group (r=-0.88, p<0.0001, r=-0.48, p=0.017, respectively) with a steeper regression slope (p=0.001). In addition, a significant interaction was demonstrated between elevated baseline hs-CRP levels and CDASs for the prediction of increased risk of future PExs (p=0.02). This interaction provided an additional indicator of clinical disease activity and added another dimension to the prior year PExs frequency phenotype to identify patients at increased risk for future PExs., Conclusion: Stable CF patients with elevated baseline hs-CRP (≥ 5.2 mg/L) demonstrated worse clinical disease activity and QOL scores at a given level of disease severity (FEV1 % predicted). Elevated baseline hs-CRP values combined with clinical disease activity scores are associated with increased risk for future IV-treated PExs even in those with mild clinical disease activity scores.- Published
- 2016
- Full Text
- View/download PDF
189. Neutrophils mediate airway hyperresponsiveness after chlorine-induced airway injury in the mouse.
- Author
-
McGovern TK, Goldberger M, Allard B, Farahnak S, Hamamoto Y, O'Sullivan M, Hirota N, Martel G, Rousseau S, and Martin JG
- Subjects
- Active Transport, Cell Nucleus, Animals, Asthma, Occupational chemically induced, Clodronic Acid administration & dosage, Cytokines metabolism, Granulocytes immunology, Lung immunology, Lung pathology, Male, Mice, Inbred BALB C, NF-E2-Related Factor 2 metabolism, Oxidative Stress, Up-Regulation, Asthma, Occupational immunology, Chlorine toxicity, Neutrophils immunology
- Abstract
Chlorine gas (Cl2) inhalation causes oxidative stress, airway epithelial damage, airway hyperresponsiveness (AHR), and neutrophilia. We evaluated the effect of neutrophil depletion on Cl2-induced AHR and its effect on the endogenous antioxidant response, and if eosinophils or macrophages influence Cl2-induced AHR. We exposed male Balb/C mice to 100 ppm Cl2 for 5 minutes. We quantified inflammatory cell populations in bronchoalveolar lavage (BAL), the antioxidant response in lung tissue by quantitative PCR, and nuclear factor (erythroid-derived 2)-like 2 (NRF2) nuclear translocation by immunofluorescence. In vitro, NRF2 nuclear translocation in response to exogenous hypochlorite was assessed using a luciferase assay. Anti-granulocyte receptor-1 antibody or anti-Ly6G was used to deplete neutrophils. The effects of neutrophil depletion on IL-13 and IL-17 were measured by ELISA. Eosinophils and macrophages were depleted using TRFK5 or clodronate-loaded liposomes, respectively. AHR was evaluated with the constant-phase model in response to inhaled aerosolized methacholine. Our results show that Cl2 exposure induced neutrophilia and increased expression of NRF2 mRNA, superoxide dismutase-1, and heme-oxygenase 1. Neutrophil depletion abolished Cl2-induced AHR in large conducting airways and prevented increases in antioxidant gene expression and NRF2 nuclear translocation. Exogenous hypochlorite administration resulted in increased NRF2 nuclear translocation in vitro. After Cl2 exposure, neutrophils occupied 22 ± 7% of the luminal space in large airways. IL-17 in BAL was increased after Cl2, although this effect was not prevented by neutrophil depletion. Neither depletion of eosinophils nor macrophages prevented Cl2-induced AHR. Our data suggest the ability of neutrophils to promote Cl2-induced AHR is dependent on increases in oxidative stress and occupation of luminal space in large airways.
- Published
- 2015
- Full Text
- View/download PDF
190. Gram negative bacteria increase non-small cell lung cancer metastasis via Toll-like receptor 4 activation and mitogen-activated protein kinase phosphorylation.
- Author
-
Chow SC, Gowing SD, Cools-Lartigue JJ, Chen CB, Berube J, Yoon HW, Chan CH, Rousseau MC, Bourdeau F, Giannias B, Roussel L, Qureshi ST, Rousseau S, and Ferri LE
- Subjects
- Animals, Cell Adhesion, Cell Line, Tumor, Humans, Male, Mice, Mice, Inbred C57BL, Phosphorylation, Carcinoma, Non-Small-Cell Lung secondary, Escherichia coli pathogenicity, Lung Neoplasms pathology, Mitogen-Activated Protein Kinases metabolism, Toll-Like Receptor 4 physiology
- Abstract
Surgery is required for the curative treatment of lung cancer but is associated with high rates of postoperative pneumonias predominantly caused by gram negative bacteria. Recent evidence suggests that these severe infectious complications may decrease long term survival after hospital discharge via cancer recurrence, but the mechanism is unclear. Lung cancer cells have recently been demonstrated to express Toll-like receptors (TLR) that mediate pathogen recognition. We hypothesized that incubation of non-small cell lung cancer (NSCLC) cells with heat-inactivated Escherichia coli can augment cancer cell adhesion, migration and metastasis via TLR4 signaling. Incubation of murine and human NSCLC cells with E. coli increased in vitro cell adhesion to collagen I, collagen IV and fibronectin, and enhanced in vitro migration. Using hepatic intravital microscopy, we demonstrated that NSCLC cells have increased in vivo adhesion to hepatic sinusoids after coincubation with gram negative bacteria. These enhanced cell adhesion and migration phenotypes following incubation with E. coli were attenuated at three levels: inhibition of TLR4 (Eritoran), p38 MAPK (BIRB0796) and ERK1/2 phosphorylation (PD184352). Incubation of murine NSCLC cells in vitro with E. coli prior to intrasplenic injection significantly augmented formation of in vivo hepatic metastases 2 weeks later. This increase was abrogated by NSCLC TLR4 blockade using Eritoran. TLR4 represents a potential therapeutic target to help prevent severe postoperative infection driven cancer metastasis., (© 2014 UICC.)
- Published
- 2015
- Full Text
- View/download PDF
191. NLRX1 prevents mitochondrial induced apoptosis and enhances macrophage antiviral immunity by interacting with influenza virus PB1-F2 protein.
- Author
-
Jaworska J, Coulombe F, Downey J, Tzelepis F, Shalaby K, Tattoli I, Berube J, Rousseau S, Martin JG, Girardin SE, McCullers JA, and Divangahi M
- Subjects
- Animals, Cell Line, Tumor, Humans, Inflammation, Influenza A virus physiology, Macrophages metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Mitochondrial Proteins metabolism, Protein Binding, Protein Structure, Tertiary, Virus Replication, Apoptosis, Influenza A virus immunology, Macrophages immunology, Mitochondria metabolism, Mitochondrial Proteins genetics, Viral Proteins metabolism
- Abstract
To subvert host immunity, influenza A virus (IAV) induces early apoptosis in innate immune cells by disrupting mitochondria membrane potential via its polymerase basic protein 1-frame 2 (PB1-F2) accessory protein. Whether immune cells have mechanisms to counteract PB1-F2-mediated apoptosis is currently unknown. Herein, we define that the host mitochondrial protein nucleotide-binding oligomerization domain-like receptor (NLR)X1 binds to viral protein PB1-F2, preventing IAV-induced macrophage apoptosis and promoting both macrophage survival and type I IFN signaling. We initially observed that Nlrx1-deficient mice infected with IAV exhibited increased pulmonary viral replication, as well as enhanced inflammatory-associated pulmonary dysfunction and morbidity. Analysis of the lungs of IAV-infected mice revealed markedly enhanced leukocyte recruitment but impaired production of type I IFN in Nlrx1(-/-) mice. Impaired type I IFN production and enhanced viral replication was recapitulated in Nlrx1(-/-) macrophages and was associated with increased mitochondrial mediated apoptosis. Through gain- and loss-of-function strategies for protein interaction, we identified that NLRX1 directly bound PB1-F2 in the mitochondria of macrophages. Using a recombinant virus lacking PB1-F2, we confirmed that deletion of PB1-F2 abrogated NLRX1-dependent macrophage type I IFN production and apoptosis. Thus, our results demonstrate that NLRX1 acts as a mitochondrial sentinel protecting macrophages from PB1-F2-induced apoptosis and preserving their antiviral function. We further propose that NLRX1 is critical for macrophage immunity against IAV infection by sensing the extent of viral replication and maintaining a protective balance between antiviral immunity and excessive inflammation within the lungs.
- Published
- 2014
- Full Text
- View/download PDF
192. The protein kinases TPL2 and EGFR contribute to ERK1/ERK2 hyperactivation in CFTRΔF508-expressing airway epithelial cells exposed to Pseudomonas aeruginosa.
- Author
-
Martel G, Roussel L, and Rousseau S
- Subjects
- Cell Line, Cystic Fibrosis genetics, Enzyme Activation, Humans, MAP Kinase Signaling System, Mitogen-Activated Protein Kinase 1 biosynthesis, Mitogen-Activated Protein Kinase 3 biosynthesis, Pseudomonas Infections genetics, Respiratory Mucosa microbiology, Sequence Deletion, Cystic Fibrosis enzymology, Cystic Fibrosis microbiology, Cystic Fibrosis Transmembrane Conductance Regulator genetics, ErbB Receptors metabolism, MAP Kinase Kinase Kinases metabolism, Proto-Oncogene Proteins metabolism, Pseudomonas Infections enzymology, Pseudomonas aeruginosa, Respiratory Mucosa enzymology
- Abstract
Excessive inflammation and Pseudomonas aeruginosa infection are two major characteristics of cysticfibrosis (CF) lung disease. In this manuscript, we describe a novel mechanism of ERK1/ERK2 activationand CXCL8 expression in airway epithelial cells (AECs) lacking functional CFTR. In both non-CF and CFAECs, the protein kinase TPL2 is required for ERK1/ERK2 MAPK activation. However, we have found that EGFR is strongly phosphorylated in the airway epithelium of CF lung and contributes to ERK1/ERK2 MAPK activation in CF AECs exposed to P. aeruginosa diffusible material (PsaDM). Moreover, PsaDM stimulates the expression of the EGFR pro-ligand HB-EGF more strongly, and in a sustained manner, in CF AECs compared to non-CF cells. Finally, although both non-CF and CF AECs expresses CXCL8 in response to PsaDM, the levels of CXCL8 are higher and EGFR plays a more important role in regulating CXCL8 synthesis in CF AECs. Together, our finding shows that in addition to the TLR-mediated TPL2 activation of ERK1/ERK2, an additional pathway contributing to ERK1/ERK2 activation is triggered by infection of CF AECs: the EGFR signaling pathway. This second pathway may contribute to excessive inflammation observed in CF.
- Published
- 2013
193. Lipopolysaccharide-induced toll-like receptor 4 signaling enhances the migratory ability of human esophageal cancer cells in a selectin-dependent manner.
- Author
-
Rousseau MC, Hsu RY, Spicer JD, McDonald B, Chan CH, Perera RM, Giannias B, Chow SC, Rousseau S, Law S, and Ferri LE
- Subjects
- Animals, Cell Adhesion drug effects, Cell Movement drug effects, Fibronectins metabolism, Humans, Liver pathology, Male, Mice, Mice, Inbred C57BL, Toll-Like Receptor 4 drug effects, p38 Mitogen-Activated Protein Kinases physiology, Esophageal Neoplasms pathology, Lipopolysaccharides pharmacology, Selectins physiology, Signal Transduction drug effects, Toll-Like Receptor 4 physiology
- Abstract
Background: Esophageal cancer is an aggressive malignancy, and emerging data suggest that postoperative infections may promote cancer progression. Systemic exposure to lipopolysaccharide (LPS), a Gram-negative bacterial antigen involved in such infections, has been shown to increase cancer cell adhesion to the hepatic sinusoids in vivo. We investigated the direct impact of LPS on the migratory ability of esophageal cancer cells via the LPS receptor toll-like receptor 4 (TLR4)., Methods: Human esophageal squamous carcinoma cell lines and immortalized normal esophageal mucosa cells were tested for TLR4 surface expression by reverse transcription polymerase chain reaction (RT-PCR) and flow cytometry. TLR4 signaling in response to LPS stimulation was tested in these cells by measuring p38 MAP kinase phosphorylation on Western blot. The impact of TLR4 signaling was measured by static adhesion assays in vitro and on early in vivo migration by intravital microscopy of the liver., Results: Upon LPS stimulation, phosphorylation of p38 was detected in the human esophageal cancer cells HKESC-2. Also, LPS-stimulated HKESC-2 cells showed a twofold increased adhesion to fibronectin and to hepatic sinusoidal endothelium. These effects were abolished by TLR4 inhibition using the small-molecule inhibitor eritoran. Adhesion to fibronectin and hepatic sinusoidal endothelium was also diminished by blockade of p38 phosphorylation and inhibitors of selectin-selectin ligand binding., Conclusion: LPS can increase the migratory ability of human esophageal cancer cells by increasing their adhesive properties through TLR4 signaling and selectin ligands. TLR4, p38, and selectin blockade may therefore prove to be a new therapeutic strategy for this aggressive malignancy., (Copyright © 2013 Mosby, Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
194. IL-33 is expressed in epithelia from patients with cystic fibrosis and potentiates neutrophil recruitment.
- Author
-
Roussel L, Farias R, and Rousseau S
- Subjects
- Humans, Interleukin-33, Interleukins genetics, Pseudomonas aeruginosa immunology, RNA, Messenger metabolism, Respiratory Mucosa immunology, Cystic Fibrosis immunology, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Interleukins immunology, Neutrophil Infiltration
- Published
- 2013
- Full Text
- View/download PDF
195. Steroid-insensitive ERK1/2 activity drives CXCL8 synthesis and neutrophilia by airway smooth muscle.
- Author
-
Robins S, Roussel L, Schachter A, Risse PA, Mogas AK, Olivenstein R, Martin JG, Hamid Q, and Rousseau S
- Subjects
- Adult, Aged, Aged, 80 and over, Anti-Asthmatic Agents therapeutic use, Asthma drug therapy, Asthma pathology, Cells, Cultured, Dual Specificity Phosphatase 1 biosynthesis, Female, Glucocorticoids therapeutic use, Humans, Male, Middle Aged, Muscle, Smooth drug effects, Muscle, Smooth metabolism, Myocytes, Smooth Muscle drug effects, Myocytes, Smooth Muscle metabolism, Severity of Illness Index, Tumor Necrosis Factor-alpha pharmacology, Up-Regulation, Young Adult, Asthma metabolism, Interleukin-8 biosynthesis, MAP Kinase Signaling System, Neutrophils metabolism
- Abstract
Severe or refractory asthma affects 5 to 15% of all patients with asthma, but is responsible for more than half of the health burden associated with the disease. Severe asthma is characterized by a dramatic increase in smooth muscle and airway inflammation. Although glucocorticoids are the mainstay of treatment in asthma, they are unable to fully control the disease in individuals with severe asthma. We found that airway smooth muscle cells (ASMCs) from individuals with severe asthma showed elevated activities of the ERK1/ERK2 and p38 MAPK pathways despite treatment with oral and inhaled glucocorticoids, which increased the expression of DUSP1, a phosphatase shown to limit p38 MAPK activity. In ex vivo ASMCs, TNF-α but not IL-17A induced expression of the neutrophil chemoattractant CXCL8. Moreover, TNF-α led to up-regulation of the ERK1/ERK2 and p38 MAPKs pathways, with only the latter being sensitive to pretreatment with the glucocorticoid dexamethasone. In contrast to epithelial and endothelial cells, TNF-α-stimulated CXCL8 synthesis was dependent on ERK1/ERK2 but not on p38 MAPK. Moreover, suppressing ERK1/ERK2 activation prevented neutrophil recruitment by ASMCs, whereas suppressing p38 MAPK activity had no impact. Taken together, these results highlight the ERK1/ERK2 MAPK cascade as a novel and attractive target in severe asthma because the activation of this pathway is insensitive to the action of glucocorticoids and is involved in neutrophil recruitment, contributing the to inflammation seen in the disease.
- Published
- 2011
- Full Text
- View/download PDF
196. Steroids and extracellular signal-regulated kinase 1/2 activity suppress activating transcription factor 3 expression in patients with severe asthma.
- Author
-
Roussel L, Robins S, Schachter A, Bérubé J, Hamid Q, and Rousseau S
- Subjects
- Adult, Asthma genetics, Base Sequence, Case-Control Studies, Cell Line, DNA Primers genetics, Gene Expression drug effects, Humans, RNA, Messenger genetics, RNA, Messenger metabolism, Toll-Like Receptor 3 agonists, Toll-Like Receptor 9 agonists, Tumor Necrosis Factor-alpha pharmacology, Activating Transcription Factor 3 genetics, Activating Transcription Factor 3 metabolism, Asthma drug therapy, Asthma metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Glucocorticoids pharmacology
- Published
- 2011
- Full Text
- View/download PDF
197. LPS-induced TLR4 signaling in human colorectal cancer cells increases beta1 integrin-mediated cell adhesion and liver metastasis.
- Author
-
Hsu RY, Chan CH, Spicer JD, Rousseau MC, Giannias B, Rousseau S, and Ferri LE
- Subjects
- Animals, Blotting, Western, Cell Adhesion drug effects, Cell Separation, Colorectal Neoplasms metabolism, Flow Cytometry, HT29 Cells, Humans, Integrin beta1 metabolism, Liver Neoplasms secondary, Lymphocyte Antigen 96 metabolism, Mice, Mice, Nude, Microscopy, Fluorescence, Neoplasm Metastasis pathology, Signal Transduction drug effects, Signal Transduction physiology, Antigens, Bacterial pharmacology, Colorectal Neoplasms pathology, Lipopolysaccharides pharmacology, Liver Neoplasms metabolism, Toll-Like Receptor 4 metabolism
- Abstract
Infectious complications resulting from resection of colorectal cancer (CRC) elevates the risk of cancer recurrence and metastasis, but the reason for this risk relationship is unknown. Defining the mechanisms responsible may offer opportunities to improve outcomes in a majority of patients whose tumors are resected as part of their therapy. The complex formed between Toll receptor TLR4 and myeloid differentiation factor MD2 defines a major cell surface receptor for lipopolysaccharide (LPS), a gram-negative bacterial antigen that has been implicated in infectious complications after CRC resection. As the TLR4/MD2 complex is expressed on CRC cells, we hypothesized that LPS may promote liver metastasis in CRC by stimulating TLR4 signaling. In support of this hypothesis, we report here that LPS enhances liver metastasis of human CRC cells that express TLR4/MD2 after intrasplenic graft of immunocompromised nude mice. Compared with TLR4 nonexpressing, nonmetastatic CRC cells, we observed increased in vitro adherence to different extracellular matrices and human umbilical vein endothelial cells (HUVEC). Furthermore, we observed an increased likelihood of in vivo capture within hepatic sinusoids after LPS treatment. No differences were apparent in phosphorylation of p38 and MAPK isoforms, but in metastatic CRC cells expressing surface TLR4 treatment with LPS increased Ser473 phosphorylation of AKT kinase. We showed that enhanced adherence elicited by LPS in these cells could be blocked at three different levels, using Eritoran (TLR4 small molecule antagonist), PI-103 (PI3K inhibitor), or anti-β1 integrin blocking antibodies. Taken together, the results indicate that stimulation of the TLR4/MD2 complex by LPS activates PI3K/AKT signaling and promotes downstream β1 integrin function, thereby increasing the adhesiveness and metastatic capacity of CRC cells. Our findings suggest that inhibiting LPS-induced TLR4 signaling could improve therapeutic outcomes by preventing cancer metastasis during the perioperative period of CRC resection., (©2011 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
198. Inhibition of SAPK2a/p38 prevents hnRNP A0 phosphorylation by MAPKAP-K2 and its interaction with cytokine mRNAs.
- Author
-
Rousseau S, Morrice N, Peggie M, Campbell DG, Gaestel M, and Cohen P
- Subjects
- Animals, Chemokine CXCL2, Enzyme Inhibitors pharmacology, Imidazoles pharmacology, Intracellular Signaling Peptides and Proteins, Mice, Monokines genetics, Phosphorylation, Pyridines pharmacology, Tumor Necrosis Factor-alpha genetics, p38 Mitogen-Activated Protein Kinases, Heterogeneous-Nuclear Ribonucleoproteins metabolism, Mitogen-Activated Protein Kinases metabolism, Protein Serine-Threonine Kinases metabolism, RNA, Messenger metabolism
- Abstract
Lipopolysaccharide (LPS) stimulates production of inflammatory mediators, partly by stabilizing [interleukin-6 (IL-6), cyclooxygenase 2 (COX-2)] and/or stimulating translation [tumour necrosis factor-alpha (TNF-alpha)] of their mRNAs. Such regulation depends on AU-rich elements (AREs) within the 3'-untranslated regions and is partially suppressed by SB 203580 (which inhibits SAPK2a/p38). The LPS-induced production of TNF-alpha and IL-6 is suppressed in MAPKAP-K2-deficient mice (a kinase activated by SAPK2a/p38). Here, we identify 18 macrophage proteins that bind to AREs and show that hnRNP A0 is a major substrate for MAPKAP-K2 in this fraction. MAPKAP-K2 phosphorylated hnRNP A0 at Ser84 in vitro and this residue became phosphorylated in LPS-stimulated cells. Phosphorylation was prevented by SB 203580 and suppressed in macrophages derived from MAPKAP-K2-deficient mice. The mRNAs encoding TNF-alpha, COX-2 and macrophage inflammatory protein-2 (MIP-2) bound to hnRNP A0 in LPS-stimulated macrophages, an interaction prevented by SB 203580. The LPS-induced stabilization of MIP-2 mRNA and production of MIP-2 protein were abolished when macrophages were incubated with SB 203580 plus PD 184352 (which inhibits the classical MAP kinase cascade). Our data suggest that LPS-induced binding of hnRNP A0 to AREs may contribute to the post-transcriptional regulation of specific mRNAs.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.