80 results on '"Cougoule C"'
Search Results
2. Antiviral and Anti-Inflammatory Activities of Fluoxetine in a SARS-CoV-2 Infection Mouse Model
- Author
-
Péricat, D., primary, Leon-Icaza, S. A., additional, Sánchez-Rico, M., additional, Mühle, C., additional, Zoicas, I., additional, Schumacher, F., additional, Planès, R., additional, Mazars, R., additional, Gros, G., additional, Carpinteiro, A., additional, Becker, K. A., additional, Izopet, J., additional, Strub-Wourgaft, N., additional, Sjö, P., additional, Neyrolles, O., additional, Kleuser, B., additional, Limosin, F., additional, Gulbins, E., additional, Kornhuber, J., additional, Meunier, E., additional, Hoertel, N., additional, and Cougoule, C., additional
- Published
- 2023
- Full Text
- View/download PDF
3. Antidepressant Use and Its Association with 28-Day Mortality in Inpatients with SARS-CoV-2: Support for the FIASMA Model against COVID-19
- Author
-
Hoertel, N., primary, Sanchez-Rico, M., additional, Kornhuber, J., additional, Gulbins, E., additional, Reiersen, A., additional, Lenze, E., additional, Fritz, B. A., additional, Jalali, F., additional, Mills, E., additional, Cougoule, C., additional, Carpinteiro, A., additional, Mühle, C., additional, Becker-Flegler, K. A., additional, Boulware, D. R., additional, Blanco, C., additional, Alvarado, J. M., additional, Strub-Wourgaft, N., additional, Lemogne, C., additional, and Limosin, F., additional
- Published
- 2023
- Full Text
- View/download PDF
4. Association between benzodiazepine receptor agonist use and mortality in patients hospitalised for COVID-19: a multicentre observational study
- Author
-
Hoertel, N., Sánchez Rico, M., Gulbins, E., Kornhuber, J., Vernet, R., Beeker, N., Neuraz, A., Blanco, C., Olfson, M., Airagnes, G., Lemogne, C., Alvarado Izquierdo, Jesús María, Arnaout, M., Cougoule, C., Meneton, P., Limosin, F., Hoertel, N., Sánchez Rico, M., Gulbins, E., Kornhuber, J., Vernet, R., Beeker, N., Neuraz, A., Blanco, C., Olfson, M., Airagnes, G., Lemogne, C., Alvarado Izquierdo, Jesús María, Arnaout, M., Cougoule, C., Meneton, P., and Limosin, F.
- Abstract
CRUE-CSIC (Acuerdos Transformativos 2022), Aims To examine the association between benzodiazepine receptor agonist (BZRA) use and mortality in patients hospitalised for coronavirus disease 2019 (COVID-19). Methods A multicentre observational study was performed at Greater Paris University hospitals. The sample involved 14 381 patients hospitalised for COVID-19. A total of 686 (4.8%) inpatients received a BZRA at hospital admission at a mean daily diazepam-equivalent dose of 19.7 mg (standard deviation (S.D.) = 25.4). The study baseline was the date of admission, and the primary endpoint was death. We compared this endpoint between patients who received BZRAs and those who did not in time-to-event analyses adjusted for sociodemographic characteristics, medical comorbidities and other medications. The primary analysis was a Cox regression model with inverse probability weighting (IPW). Results Over a mean follow-up of 14.5 days (S.D. = 18.1), the primary endpoint occurred in 186 patients (27.1%) who received BZRAs and in 1134 patients (8.3%) who did not. There was a significant association between BZRA use and increased mortality both in the crude analysis (hazard ratio (HR) = 3.20; 95% confidence interval (CI) = 2.74–3.74; p < 0.01) and in the IPW analysis (HR = 1.61; 95% CI = 1.31–1.98, p < 0.01), with a significant dose-dependent relationship (HR = 1.55; 95% CI = 1.08–2.22; p = 0.02). This association remained significant in sensitivity analyses. Exploratory analyses indicate that most BZRAs may be associated with an increased mortality among patients hospitalised for COVID-19, except for diazepam, which may be associated with a reduced mortality compared with any other BZRA treatment. Conclusions BZRA use may be associated with an increased mortality among patients hospitalised for COVID-19, suggesting the potential benefit of decreasing dose or tapering off gradually these medications when possible., Depto. de Psicobiología y Metodología en Ciencias del Comportamiento, Fac. de Psicología, TRUE, pub
- Published
- 2022
5. Human NLRP1 is a sensor of pathogenic coronavirus 3CL proteases in lung epithelial cells
- Author
-
Reversade, Bruno, Planes, R.; Pinilla, M.; Santoni, K.; Hessel, A.; Passemar, C.; Lay, K.; Paillette, P.; Valadao, A.C.; Robinson, K.S.; Bastard, P.; Lam, N.; Fadrique, R.; Rossi, I.; Pericat, D.; Bagayoko, S.; Leon-Icaza, S.A.; Rombouts, Y.; Perouzel, E.; Tiraby, M.; COVID Human Genetic Effort; Zhang, Q.; Cicuta, P.; Jouanguy, E.; Neyrolles, O.; Bryant, C.E.; Floto, A.R.; Goujon, C.; Lei, F.Z.; Martin-Blondel, G.; Silva, S.; Casanova, J.L.; Cougoule, C.; Marcoux, J.; Ravet, E.; Meunier, E., School of Medicine, Reversade, Bruno, Planes, R.; Pinilla, M.; Santoni, K.; Hessel, A.; Passemar, C.; Lay, K.; Paillette, P.; Valadao, A.C.; Robinson, K.S.; Bastard, P.; Lam, N.; Fadrique, R.; Rossi, I.; Pericat, D.; Bagayoko, S.; Leon-Icaza, S.A.; Rombouts, Y.; Perouzel, E.; Tiraby, M.; COVID Human Genetic Effort; Zhang, Q.; Cicuta, P.; Jouanguy, E.; Neyrolles, O.; Bryant, C.E.; Floto, A.R.; Goujon, C.; Lei, F.Z.; Martin-Blondel, G.; Silva, S.; Casanova, J.L.; Cougoule, C.; Marcoux, J.; Ravet, E.; Meunier, E., and School of Medicine
- Abstract
Inflammation observed in SARS-CoV-2-infected patients suggests that inflammasomes, proinflammatory intracellular complexes, regulate various steps of infection. Lung epithelial cells express inflammasome-forming sensors and constitute the primary entry door of SARS-CoV-2. Here, we describe that the NLRP1 in-flammasome detects SARS-CoV-2 infection in human lung epithelial cells. Specifically, human NLRP1 is cleaved at the Q333 site by multiple coronavirus 3CL proteases, which triggers inflammasome assembly and cell death and limits the production of infectious viral particles. Analysis of NLRP1-associated pathways unveils that 3CL proteases also inactivate the pyroptosis executioner Gasdermin D (GSDMD). Subsequently, caspase-3 and GSDME promote alternative cell pyroptosis. Finally, analysis of pyroptosis markers in plasma from COVID-19 patients with characterized severe pneumonia due to autoantibodies against, or inborn errors of, type I interferons (IFNs) highlights GSDME/caspase-3 as potential markers of disease severity. Overall, our findings identify NLRP1 as a sensor of SARS-CoV-2 infection in lung epithelia., Fondation pour la Recherche Med-icale (F.R.M.); European Union (EU); Horizon 2020; ERC StG (INFLAME); ERC StG (ANTIViR); French Ministry of Health; Goupe-ment Interregional de Recherche Clinique et d’Innovation Sud-Ouest Outre-Mer (PHRCI 2020 IMMUNOMARK-COV); LABEX; CIFRE PhD Fellowship; Investissement d'Avenir and foundation Bettencourt; InvivoGen; Mali Ministry of Education; Vaincre La Mucoviscidose (VLM); InvivoGen
- Published
- 2022
6. Association between benzodiazepine receptor agonist use and mortality in patients hospitalised for COVID-19: a multicentre observational study
- Author
-
Hoertel, N., primary, Sánchez-Rico, M., additional, Gulbins, E., additional, Kornhuber, J., additional, Vernet, R., additional, Beeker, N., additional, Neuraz, A., additional, Blanco, C., additional, Olfson, M., additional, Airagnes, G., additional, Lemogne, C., additional, Alvarado, J. M., additional, Arnaout, M., additional, Cougoule, C., additional, Meneton, P., additional, and Limosin, F., additional
- Published
- 2022
- Full Text
- View/download PDF
7. Characterization of the resolution phase of inflammation in the thioglycollate-induced peritonitis model: 1.16
- Author
-
Lastrucci, C., Cougoule, C., Ballif, V., Behar, A., Saati, Al T., Dubourdeau, M., and Maridonneau-Parini, I.
- Published
- 2013
8. Overview of leukocyte 3D migration: necessary role of podosomes for mesenchymal 3D migration: 155
- Author
-
Cougoule, C., Van-Goethem, E., Le-Cabec, V., Lafouresse, F., Dupré, L., Lugo-Villarino, G., Neyrolles, O., Mehraj, V., Mège, J. L., Behar, A., and Maridonneau-Parini, I.
- Published
- 2012
9. Mechanisms of macrophage migration in 3D environments, role of podosomes: 125
- Author
-
Maridonneau-Parini, I., Labernadie, A., Lastrucci, C., Guiet, R., Charrière, G., Le Cabec, V., and Cougoule, C.
- Published
- 2012
10. Hck is involved in 3D macrophage migration in vitro and in vivo: 105
- Author
-
Cougoule, C., Laviolette, N., van Goethem, E., Poincloux, R., Lowell, C. A., Mège, J. L., Le Cabec, V., and Maridonneau-Parini, I.
- Published
- 2010
11. The phagocyte-specific tyrosine kinase Hck is involved in lysosome propelling, podosome formation and transtissular migration: 47
- Author
-
Vincent, C., Labrousse, A., Poincloux, R., Cougoule, C., Le Cabec, V., and Maridonneau-Parini, I.
- Published
- 2007
12. The two isoforms of Hck, a phagocyte-specific Src family tyrosine kinase, are necessary for cell migration through an extracellular matrix: 94
- Author
-
Poincloux, R., Cougoule, C., Bordier, C., Daubon, T., Cabec, V. Le, and Maridonneau-Parini, I.
- Published
- 2006
13. Relocation of STAT5 to podosomes when activated by the oncogenic form of the tyrosine kinase Hck: 122
- Author
-
Poincloux, R., Cougoule, C., Daubon, T., Maridonneau-Parini, I., and Cabec, V. Le
- Published
- 2005
14. The phagocyte-specific Src tyrosine kinase, Hck, requires the synergic action of its lysosome- and plasma membrane-associated isoforms to induce migratory and tumorigenic abilities of NIH3T3 cells
- Author
-
Le Cabec, V., Poincloux, R., Cougoule, C., and Maridonneau-Parini, I.
- Published
- 2004
15. EspF of enteropathogenic Escherichia coli binds sorting nexin 9
- Author
-
Marchès, O, Batchelor, M, Shaw, RK, Patel, A, Cummings, N, Nagai, T, Sasakawa, C, Carlsson, Sven R, Lundmark, Richard, Cougoule, C, Caron, E, Knutton, S, Connerton, I, Frankel, G, Marchès, O, Batchelor, M, Shaw, RK, Patel, A, Cummings, N, Nagai, T, Sasakawa, C, Carlsson, Sven R, Lundmark, Richard, Cougoule, C, Caron, E, Knutton, S, Connerton, I, and Frankel, G
- Published
- 2006
16. Phagocytosis, an alternative model system for the study of cell adhesion
- Author
-
COUGOULE, C, primary, WIEDEMANN, A, additional, LIM, J, additional, and CARON, E, additional
- Published
- 2004
- Full Text
- View/download PDF
17. Human NLRP1 is a sensor of pathogenic coronavirus 3CL proteases in lung epithelial cells
- Author
-
Rémi Planès, Miriam Pinilla, Karin Santoni, Audrey Hessel, Charlotte Passemar, Kenneth Lay, Perrine Paillette, Ana-Luiza Chaves Valadão, Kim Samirah Robinson, Paul Bastard, Nathaniel Lam, Ricardo Fadrique, Ida Rossi, David Pericat, Salimata Bagayoko, Stephen Adonai Leon-Icaza, Yoann Rombouts, Eric Perouzel, Michèle Tiraby, Qian Zhang, Pietro Cicuta, Emmanuelle Jouanguy, Olivier Neyrolles, Clare E. Bryant, Andres R. Floto, Caroline Goujon, Franklin Zhong Lei, Guillaume Martin-Blondel, Stein Silva, Jean-Laurent Casanova, Céline Cougoule, Bruno Reversade, Julien Marcoux, Emmanuel Ravet, Etienne Meunier, Institut de pharmacologie et de biologie structurale (IPBS), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS), InvivoGen Europe, Institut de Recherche en Infectiologie de Montpellier (IRIM), Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM), Molecular Immunity Unit [Cambridge, UK] (Department of Medicine), University of Cambridge [UK] (CAM), Institute of Medical Biology [Singapore Singapore], Genome Institute of Singapore (GIS), Agency for science, technology and research [Singapore] (A*STAR), Human genetics of infectious diseases : Mendelian predisposition (Equipe Inserm U1163), Imagine - Institut des maladies génétiques (IHU) (Imagine - U1163), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University [New York], Department of Veterinary Medicine, University of Cambridge, Cambridge, UK, School of Clinical Medicine, Cavendish Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Lee Kong Chian School of Medicine, Nanyang Technological University [Singapour], Skin Research Institute of Singapore [Singapore, Singapore] (SRIS / A*STAR), Service Maladies infectieuses et tropicales [CHU Toulouse], Pôle Inflammation, infection, immunologie et loco-moteur [CHU Toulouse] (Pôle I3LM Toulouse), Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)-Centre Hospitalier Universitaire de Toulouse (CHU Toulouse), Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Service de réanimation infantile [CHU Purpan], Centre Hospitalier Universitaire de Toulouse (CHU Toulouse), Howard Hughes Medical Institute [New York] (HHMI), Howard Hughes Medical Institute (HHMI)-New York University School of Medicine, NYU System (NYU)-NYU System (NYU)-Rockefeller University [New York]-Columbia University Irving Medical Center (CUIMC), Institute of Molecular and Cell Biology, National University of Singapore (NUS)-Agency for science, technology and research [Singapore] (A*STAR), Yong Loo Lin School of Medicine [Singapore], Koç University, This project was funded by grants from the Fondation pour la Recherche Médicale (F.R.M.) and the ERC StG (INFLAME) to E.M., the ERC StG (ANTIViR) to C.G., and the French Ministry of Health with the participation of the Groupement Interrégional de Recherche Clinique et d’Innovation Sud-Ouest Outre-Mer (PHRCI 2020 IMMUNOMARK-COV) to G.-M.B. The ASB3 structure is supported by LABEX, Investissement d’Avenir and foundation Bettencourt grants to O.N. M.P. and R.P. were, respectively, funded by a CIFRE PhD fellowship and a research grant from InvivoGen. S.B. is supported by a PhD fellowship from Mali Ministry of Education and from the FRM (FDT 12794)., ANR-12-BSV3-0002,B-TB,Rôle des lymphocytes B dans l'immunité et l'inflammation tuberculeuse(2012), ANR-18-CE44-0005,DCIR-TB,Etude des mécanismes impliqués dans la modulation de la signalisation par les interférons de type I et l'immunité anti-tuberculeuse par la lectine de type C DCIR(2018), Cougoule, Céline, BLANC - Rôle des lymphocytes B dans l'immunité et l'inflammation tuberculeuse - - B-TB2012 - ANR-12-BSV3-0002 - BLANC - VALID, APPEL À PROJETS GÉNÉRIQUE 2018 - Etude des mécanismes impliqués dans la modulation de la signalisation par les interférons de type I et l'immunité anti-tuberculeuse par la lectine de type C DCIR - - DCIR-TB2018 - ANR-18-CE44-0005 - AAPG2018 - VALID, Center for Reproductive Medicine, ACS - Heart failure & arrhythmias, Amsterdam Reproduction & Development, Reversade, Bruno, Planes, R., Pinilla, M., Santoni, K., Hessel, A., Passemar, C., Lay, K., Paillette, P., Valadao, A.C., Robinson, K.S., Bastard, P., Lam, N., Fadrique, R., Rossi, I., Pericat, D., Bagayoko, S., Leon-Icaza, S.A., Rombouts, Y., Perouzel, E., Tiraby, M., COVID Human Genetic Effort, Zhang, Q., Cicuta, P., Jouanguy, E., Neyrolles, O., Bryant, C.E., Floto, A.R., Goujon, C., Lei, F.Z., Martin-Blondel, G., Silva, S., Casanova, J.L., Cougoule, C., Marcoux, J., Ravet, E., Meunier, E., and School of Medicine
- Subjects
Pore Forming Cytotoxic Proteins ,Caspase 3 ,Inflammasomes ,SARS-CoV-2 ,[SDV]Life Sciences [q-bio] ,pyroptosis ,COVID-19 ,NLR Proteins ,Cell Biology ,3CL proteases ,Epithelial cells ,Gasdermins ,NLRP1 inflammasome ,Pyroptosis ,Phosphate-Binding Proteins ,epithelial cells ,[SDV] Life Sciences [q-bio] ,Humans ,Biochemistry and molecular biology ,Cell biology ,Lung ,Molecular Biology ,Coronavirus 3C Proteases ,Peptide Hydrolases - Abstract
Inflammation observed in SARS-CoV-2-infected patients suggests that inflammasomes, proinflammatory intracellular complexes, regulate various steps of infection. Lung epithelial cells express inflammasome-forming sensors and constitute the primary entry door of SARS-CoV-2. Here, we describe that the NLRP1 in-flammasome detects SARS-CoV-2 infection in human lung epithelial cells. Specifically, human NLRP1 is cleaved at the Q333 site by multiple coronavirus 3CL proteases, which triggers inflammasome assembly and cell death and limits the production of infectious viral particles. Analysis of NLRP1-associated pathways unveils that 3CL proteases also inactivate the pyroptosis executioner Gasdermin D (GSDMD). Subsequently, caspase-3 and GSDME promote alternative cell pyroptosis. Finally, analysis of pyroptosis markers in plasma from COVID-19 patients with characterized severe pneumonia due to autoantibodies against, or inborn errors of, type I interferons (IFNs) highlights GSDME/caspase-3 as potential markers of disease severity. Overall, our findings identify NLRP1 as a sensor of SARS-CoV-2 infection in lung epithelia., Fondation pour la Recherche Med-icale (F.R.M.); European Union (EU); Horizon 2020; ERC StG (INFLAME); ERC StG (ANTIViR); French Ministry of Health; Goupe-ment Interregional de Recherche Clinique et d’Innovation Sud-Ouest Outre-Mer (PHRCI 2020 IMMUNOMARK-COV); LABEX; CIFRE PhD Fellowship; Investissement d'Avenir and foundation Bettencourt; InvivoGen; Mali Ministry of Education; Vaincre La Mucoviscidose (VLM); InvivoGen
- Full Text
- View/download PDF
18. Mapping the scientific output of organoids for animal and human modeling infectious diseases: a bibliometric assessment.
- Author
-
Yan J, Monlong J, Cougoule C, Lacroix-Lamandé S, and Wiedemann A
- Subjects
- Animals, Humans, Communicable Diseases veterinary, Communicable Diseases epidemiology, Disease Models, Animal, COVID-19 epidemiology, COVID-19 virology, Organoids virology, Bibliometrics
- Abstract
The escalation of antibiotic resistance, pandemics, and nosocomial infections underscores the importance of research in both animal and human infectious diseases. Recent advancements in three-dimensional tissue cultures, or "organoids", have revolutionized the development of in vitro models for infectious diseases. Our study conducts a bibliometric analysis on the use of organoids in modeling infectious diseases, offering an in-depth overview of this field's current landscape. We examined scientific contributions from 2009 onward that focused on organoids in host‒pathogen interactions using the Web of Science Core Collection and OpenAlex database. Our analysis included temporal trends, reference aging, author, and institutional productivity, collaborative networks, citation metrics, keyword cluster dynamics, and disruptiveness of organoid models. VOSviewer, CiteSpace, and Python facilitated this analytical assessment. The findings reveal significant growth and advancements in organoid-based infectious disease research. Analysis of keywords and impactful publications identified three distinct developmental phases in this area that were significantly influenced by outbreaks of Zika and SARS-CoV-2 viruses. The research also highlights the synergistic efforts between academia and publishers in tackling global pandemic challenges. Through mostly consolidating research efforts, organoids are proving to be a promising tool in infectious disease research for both human and animal infectious disease. Their integration into the field necessitates methodological refinements for better physiological emulation and the establishment of extensive organoid biobanks. These improvements are crucial for fully harnessing the potential of organoids in understanding infectious diseases and advancing the development of targeted treatments and vaccines., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
19. Fluoxetine and Sertraline Potently Neutralize the Replication of Distinct SARS-CoV-2 Variants.
- Author
-
Thümmler L, Beckmann N, Sehl C, Soddemann M, Braß P, Bormann M, Brochhagen L, Elsner C, Hoertel N, Cougoule C, Ciesek S, Widera M, Dittmer U, Lindemann M, Horn PA, Witzke O, Kadow S, Kamler M, Gulbins E, Becker KA, and Krawczyk A
- Subjects
- Humans, Chlorocebus aethiops, Vero Cells, Animals, COVID-19 Drug Treatment, SARS-CoV-2 drug effects, Sertraline pharmacology, Fluoxetine pharmacology, Virus Replication drug effects, Antiviral Agents pharmacology, COVID-19 virology
- Abstract
The pandemic caused by SARS-CoV-2 is still a major health problem. Newly emerging variants and long-COVID-19 represent a challenge for the global health system. In particular, individuals in developing countries with insufficient health care need easily accessible, affordable and effective treatments of COVID-19. Previous studies have demonstrated the efficacy of functional inhibitors of acid sphingomyelinase against infections with various viruses, including early variants of SARS-CoV-2. This work investigated whether the acid sphingomyelinase inhibitors fluoxetine and sertraline, usually used as antidepressant molecules in clinical practice, can inhibit the replication of the former and recently emerged SARS-CoV-2 variants in vitro. Fluoxetine and sertraline potently inhibited the infection with pseudotyped virus-like particles and SARS-CoV-2 variants D614G, alpha, delta, omicron BA.1 and omicron BA.5. These results highlight fluoxetine and sertraline as priority candidates for large-scale phase 3 clinical trials at different stages of SARS-CoV-2 infections, either alone or in combination with other medications.
- Published
- 2024
- Full Text
- View/download PDF
20. EEF2-inactivating toxins engage the NLRP1 inflammasome and promote epithelial barrier disruption.
- Author
-
Pinilla M, Mazars R, Vergé R, Gorse L, Paradis M, Suire B, Santoni K, Robinson KS, Toh GA, Prouvensier L, Leon-Icaza SA, Hessel A, Péricat D, Murris M, Guet-Revillet H, Henras A, Buyck J, Ravet E, Zhong FL, Cougoule C, Planès R, and Meunier E
- Subjects
- Humans, Peptide Elongation Factor 2, Inflammasomes, Cytoplasm, NLR Proteins, Eukaryota, Cystic Fibrosis
- Abstract
Human airway and corneal epithelial cells, which are critically altered during chronic infections mediated by Pseudomonas aeruginosa, specifically express the inflammasome sensor NLRP1. Here, together with a companion study, we report that the NLRP1 inflammasome detects exotoxin A (EXOA), a ribotoxin released by P. aeruginosa type 2 secretion system (T2SS), during chronic infection. Mechanistically, EXOA-driven eukaryotic elongation factor 2 (EEF2) ribosylation and covalent inactivation promote ribotoxic stress and subsequent NLRP1 inflammasome activation, a process shared with other EEF2-inactivating toxins, diphtheria toxin and cholix toxin. Biochemically, irreversible EEF2 inactivation triggers ribosome stress-associated kinases ZAKα- and P38-dependent NLRP1 phosphorylation and subsequent proteasome-driven functional degradation. Finally, cystic fibrosis cells from patients exhibit exacerbated P38 activity and hypersensitivity to EXOA-induced ribotoxic stress-dependent NLRP1 inflammasome activation, a process inhibited by the use of ZAKα inhibitors. Altogether, our results show the importance of P. aeruginosa virulence factor EXOA at promoting NLRP1-dependent epithelial damage and identify ZAKα as a critical sensor of virulence-inactivated EEF2., (© 2023 Pinilla et al.)
- Published
- 2023
- Full Text
- View/download PDF
21. Druggable redox pathways against Mycobacterium abscessus in cystic fibrosis patient-derived airway organoids.
- Author
-
Leon-Icaza SA, Bagayoko S, Vergé R, Iakobachvili N, Ferrand C, Aydogan T, Bernard C, Sanchez Dafun A, Murris-Espin M, Mazières J, Bordignon PJ, Mazères S, Bernes-Lasserre P, Ramé V, Lagarde JM, Marcoux J, Bousquet MP, Chalut C, Guilhot C, Clevers H, Peters PJ, Molle V, Lugo-Villarino G, Cam K, Berry L, Meunier E, and Cougoule C
- Subjects
- Humans, Antioxidants, Oxidation-Reduction, Oxidative Stress, Cystic Fibrosis drug therapy, Mycobacterium abscessus
- Abstract
Mycobacterium abscessus (Mabs) drives life-shortening mortality in cystic fibrosis (CF) patients, primarily because of its resistance to chemotherapeutic agents. To date, our knowledge on the host and bacterial determinants driving Mabs pathology in CF patient lung remains rudimentary. Here, we used human airway organoids (AOs) microinjected with smooth (S) or rough (R-)Mabs to evaluate bacteria fitness, host responses to infection, and new treatment efficacy. We show that S Mabs formed biofilm, and R Mabs formed cord serpentines and displayed a higher virulence. While Mabs infection triggers enhanced oxidative stress, pharmacological activation of antioxidant pathways resulted in better control of Mabs growth and reduced virulence. Genetic and pharmacological inhibition of the CFTR is associated with better growth and higher virulence of S and R Mabs. Finally, pharmacological activation of antioxidant pathways inhibited Mabs growth, at least in part through the quinone oxidoreductase NQO1, and improved efficacy in combination with cefoxitin, a first line antibiotic. In conclusion, we have established AOs as a suitable human system to decipher mechanisms of CF-driven respiratory infection by Mabs and propose boosting of the NRF2-NQO1 axis as a potential host-directed strategy to improve Mabs infection control., Competing Interests: Julien Mazières reports grants or contracts from Astra Zeneca, Roche and Pierre Fabre; and payment or honoraria for board and expertise (personal and institution) from Merck, Astra Zeneca, BMS, MSD, Roche, Novartis, Daiichi, and Pfizer; outside the submitted work. Hans Clevers reports invention on patents related to organoid research. His full disclosure: www.uu.nl/staff/JCClevers/Additional function. The other authors have declared no competing interests., (Copyright: © 2023 Leon-Icaza 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
- 2023
- Full Text
- View/download PDF
22. Medications Modulating the Acid Sphingomyelinase/Ceramide System and 28-Day Mortality among Patients with SARS-CoV-2: An Observational Study.
- Author
-
Hoertel N, Rezaei K, Sánchez-Rico M, Delgado-Álvarez A, Kornhuber J, Gulbins E, Olfson M, Ouazana-Vedrines C, Carpinteiro A, Cougoule C, Becker KA, Alvarado JM, Limosin F, and On Behalf Of The Ap-Hp/Université Paris Cité/Inserm Covid-Research Collaboration Ap-Hp Covid Cdr Initiative And Entrepôt de Données de Santé Ap-Hp Consortium
- Abstract
Prior evidence indicates the potential central role of the acid sphingomyelinase (ASM)/ceramide system in the infection of cells with SARS-CoV-2. We conducted a multicenter retrospective observational study including 72,105 adult patients with laboratory-confirmed SARS-CoV-2 infection who were admitted to 36 AP-HP (Assistance Publique-Hôpitaux de Paris) hospitals from 2 May 2020 to 31 August 2022. We examined the association between the ongoing use of medications functionally inhibiting acid sphingomyelinase (FIASMA), which reduces the infection of cells with SARS-CoV-2 in vitro, upon hospital admission with 28-day all-cause mortality in a 1:1 ratio matched analytic sample based on clinical characteristics, disease severity and other medications (N = 9714). The univariate Cox regression model of the matched analytic sample showed that FIASMA medication use at admission was associated with significantly lower risks of 28-day mortality (HR = 0.80; 95% CI = 0.72-0.88; p < 0.001). In this multicenter observational study, the use of FIASMA medications was significantly and substantially associated with reduced 28-day mortality among adult patients hospitalized with COVID-19. These findings support the continuation of these medications during the treatment of SARS-CoV-2 infections. Randomized clinical trials (RCTs) are needed to confirm these results, starting with the molecules with the greatest effect size in the study, e.g., fluoxetine, escitalopram, and amlodipine.
- Published
- 2023
- Full Text
- View/download PDF
23. A druggable copper-signalling pathway that drives inflammation.
- Author
-
Solier S, Müller S, Cañeque T, Versini A, Mansart A, Sindikubwabo F, Baron L, Emam L, Gestraud P, Pantoș GD, Gandon V, Gaillet C, Wu TD, Dingli F, Loew D, Baulande S, Durand S, Sencio V, Robil C, Trottein F, Péricat D, Näser E, Cougoule C, Meunier E, Bègue AL, Salmon H, Manel N, Puisieux A, Watson S, Dawson MA, Servant N, Kroemer G, Annane D, and Rodriguez R
- Subjects
- Animals, Mice, Macrophages drug effects, Macrophages immunology, Macrophages metabolism, Macrophages pathology, NAD metabolism, Mitochondria drug effects, Mitochondria metabolism, Hydrogen Peroxide metabolism, Epigenesis, Genetic drug effects, Metformin analogs & derivatives, Oxidation-Reduction, Macrophage Activation drug effects, Macrophage Activation genetics, Copper metabolism, Inflammation drug therapy, Inflammation genetics, Inflammation immunology, Inflammation metabolism, Inflammation pathology, Signal Transduction drug effects, Cell Plasticity drug effects, Cell Plasticity genetics
- Abstract
Inflammation is a complex physiological process triggered in response to harmful stimuli
1 . It involves cells of the immune system capable of clearing sources of injury and damaged tissues. Excessive inflammation can occur as a result of infection and is a hallmark of several diseases2-4 . The molecular bases underlying inflammatory responses are not fully understood. Here we show that the cell surface glycoprotein CD44, which marks the acquisition of distinct cell phenotypes in the context of development, immunity and cancer progression, mediates the uptake of metals including copper. We identify a pool of chemically reactive copper(II) in mitochondria of inflammatory macrophages that catalyses NAD(H) redox cycling by activating hydrogen peroxide. Maintenance of NAD+ enables metabolic and epigenetic programming towards the inflammatory state. Targeting mitochondrial copper(II) with supformin (LCC-12), a rationally designed dimer of metformin, induces a reduction of the NAD(H) pool, leading to metabolic and epigenetic states that oppose macrophage activation. LCC-12 interferes with cell plasticity in other settings and reduces inflammation in mouse models of bacterial and viral infections. Our work highlights the central role of copper as a regulator of cell plasticity and unveils a therapeutic strategy based on metabolic reprogramming and the control of epigenetic cell states., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
24. Establishing 20S Proteasome Genetic, Translational and Post-Translational Status from Precious Biological and Patient Samples with Top-Down MS.
- Author
-
Dafun AS, Živković D, Leon-Icaza SA, Möller S, Froment C, Bonnet D, de Jesus AA, Alric L, Quaranta-Nicaise M, Ferrand A, Cougoule C, Meunier E, Burlet-Schiltz O, Ebstein F, Goldbach-Mansky R, Krüger E, Bousquet MP, and Marcoux J
- Subjects
- Animals, Humans, Cytoplasm metabolism, Mass Spectrometry methods, Cell Line, Mammals metabolism, Proteasome Endopeptidase Complex metabolism, Protein Processing, Post-Translational
- Abstract
The mammalian 20S catalytic core of the proteasome is made of 14 different subunits (α1-7 and β1-7) but exists as different subtypes depending on the cell type. In immune cells, for instance, constitutive catalytic proteasome subunits can be replaced by the so-called immuno-catalytic subunits, giving rise to the immunoproteasome. Proteasome activity is also altered by post-translational modifications (PTMs) and by genetic variants. Immunochemical methods are commonly used to investigate these PTMs whereby protein-tagging is necessary to monitor their effect on 20S assembly. Here, we present a new miniaturized workflow combining top-down and bottom-up mass spectrometry of immunopurified 20S proteasomes that analyze the proteasome assembly status as well as the full proteoform footprint, revealing PTMs, mutations, single nucleotide polymorphisms (SNPs) and induction of immune-subunits in different biological samples, including organoids, biopsies and B-lymphoblastoid cell lines derived from patients with proteasome-associated autoinflammatory syndromes (PRAAS). We emphasize the benefits of using top-down mass spectrometry in preserving the endogenous conformation of protein modifications, while enabling a rapid turnaround (1 h run) and ensuring high sensitivity (1-2 pmol) and demonstrate its capacity to semi-quantify constitutive and immune proteasome subunits., Competing Interests: The authors declare no conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
25. Drug repurposing: From the discovery of a useful pharmacological effect to making the treatment available to the patient.
- Author
-
Deplanque D, Fetro C, Ferry A, Lechat P, Beghyn T, Bernard C, Bernasconi A, Bienayme H, Cougoule C, Del Bano J, Demiot C, and Lebrun-Vignes B
- Subjects
- Humans, Public-Private Sector Partnerships, Marketing, Drug Repositioning
- Abstract
The repurposing of a medicine already on the market to a new indication could be an opportunity to respond rapidly to a therapeutic need not yet covered, particularly in the context of rare and neglected diseases, or health emergencies. However, at each stage, difficulties may arise that will prevent the repurposed drug from being provided to patients. Beyond fortuity or a systematic strategy to detect a useful pharmacological effect, the implementation of the preclinical and clinical stages is sometimes complicated by the difficulty of accessing the molecule and its pharmaceutical data. Furthermore, relevant clinical results will not always be sufficient to ensure that a marketing authorisation is obtained or that patients receive satisfactory care. In addition to describing these various obstacles, the round table provided an opportunity to put forward recommendations for overcoming them, in particular the creation of a public-private partnership structure with sufficient funding to be able to offer individualised support for projects up to and including the marketing application., (Copyright © 2022 Société française de pharmacologie et de thérapeutique. Published by Elsevier Masson SAS. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
26. Repositionnement des médicaments : de la découverte d’un effet pharmacologique utile à la mise à disposition du traitement pour le patient.
- Author
-
Deplanque D, Fetro C, Ferry A, Lechat P, Beghyn T, Bernard C, Bernasconi A, Bienayme H, Cougoule C, Del Bano J, Demiot C, and Lebrun-Vignes B
- Subjects
- Humans, Drug Repositioning, Patients
- Published
- 2023
- Full Text
- View/download PDF
27. Risk of Death in Individuals Hospitalized for COVID-19 With and Without Psychiatric Disorders: An Observational Multicenter Study in France.
- Author
-
Hoertel N, Sánchez-Rico M, de la Muela P, Abellán M, Blanco C, Leboyer M, Cougoule C, Gulbins E, Kornhuber J, Carpinteiro A, Becker KA, Vernet R, Beeker N, Neuraz A, Alvarado JM, Herrera-Morueco JJ, Airagnes G, Lemogne C, and Limosin F
- Abstract
Background: Prior research suggests that psychiatric disorders could be linked to increased mortality among patients with COVID-19. However, whether all or specific psychiatric disorders are intrinsic risk factors of death in COVID-19 or whether these associations reflect the greater prevalence of medical risk factors in people with psychiatric disorders has yet to be evaluated., Methods: We performed an observational, multicenter, retrospective cohort study to examine the association between psychiatric disorders and mortality among patients hospitalized for laboratory-confirmed COVID-19 at 36 Greater Paris University hospitals., Results: Of 15,168 adult patients, 857 (5.7%) had an ICD-10 diagnosis of psychiatric disorder. Over a mean follow-up period of 14.6 days (SD = 17.9), 326 of 857 (38.0%) patients with a diagnosis of psychiatric disorder died compared with 1276 of 14,311 (8.9%) patients without such a diagnosis (odds ratio 6.27, 95% CI 5.40-7.28, p < .01). When adjusting for age, sex, hospital, current smoking status, and medications according to compassionate use or as part of a clinical trial, this association remained significant (adjusted odds ratio 3.27, 95% CI 2.78-3.85, p < .01). However, additional adjustments for obesity and number of medical conditions resulted in a nonsignificant association (adjusted odds ratio 1.02, 95% CI 0.84-1.23, p = .86). Exploratory analyses after the same adjustments suggested that a diagnosis of mood disorders was significantly associated with reduced mortality, which might be explained by the use of antidepressants., Conclusions: These findings suggest that the increased risk of COVID-19-related mortality in individuals with psychiatric disorders hospitalized for COVID-19 might be explained by the greater number of medical conditions and the higher prevalence of obesity in this population and not by the underlying psychiatric disease., (© 2022 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
28. Oral supplementation with yeast β -glucans improves the resolution of Escherichia coli -associated inflammatory responses independently of monocyte/macrophage immune training.
- Author
-
Walachowski S, Breyne K, Secher T, Cougoule C, Guzylack-Piriou L, Meyer E, Foucras G, and Tabouret G
- Subjects
- Mice, Animals, Saccharomyces cerevisiae, Escherichia coli, Monocytes, Macrophages, Cytokines, beta-Glucans pharmacology, Yeast, Dried
- Abstract
Introduction: Confronted with the emerging threat of antimicrobial resistance, the development of alternative strategies to limit the use of antibiotics or potentiate their effect through synergy with the immune system is urgently needed. Many natural or synthetic biological response modifiers have been investigated in this context. Among them, β-glucans, a type of soluble or insoluble polysaccharide composed of a linear or branched string of glucose molecules produced by various cereals, bacteria, algae, and inferior (yeast) and superior fungi (mushrooms) have garnered interest in the scientific community, with not less than 10,000 publications over the last two decades. Various biological activities of β-glucans have been reported, such as anticancer, antidiabetic and immune-modulating effects. In vitro, yeast β-glucans are known to markedly increase cytokine secretion of monocytes/macrophages during a secondary challenge, a phenomenon called immune training., Methods: Here, we orally delivered β-glucans derived from the yeast S. cerevisiae to mice that were further challenged with Escherichia coli., Results: β-glucan supplementation protected the mice from E. coli intraperitoneal and intra-mammary infections, as shown by a lower bacterial burden and greatly diminished tissue damage. Surprisingly, this was not associated with an increased local immune response. In addition, granulocyte recruitment was transient and limited, as well as local cytokine secretion, arguing for faster resolution of the inflammatory response. Furthermore, ex-vivo evaluation of monocytes/macrophages isolated or differentiated from β-glucan-supplemented mice showed these cells to lack a trained response versus those from control mice., Conclusion: In conclusion, dietary β-glucans can improve the outcome of Escherichia coli infections and dampen tissue damages associated to excessive inflammatory response. The mechanisms associated with such protection are not necessarily linked to immune system hyper-activation or immune training., 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., (Copyright © 2022 Walachowski, Breyne, Secher, Cougoule, Guzylack-Piriou, Meyer, Foucras and Tabouret.)
- Published
- 2022
- Full Text
- View/download PDF
29. Antiviral and Anti-Inflammatory Activities of Fluoxetine in a SARS-CoV-2 Infection Mouse Model.
- Author
-
Péricat D, Leon-Icaza SA, Sanchez Rico M, Mühle C, Zoicas I, Schumacher F, Planès R, Mazars R, Gros G, Carpinteiro A, Becker KA, Izopet J, Strub-Wourgaft N, Sjö P, Neyrolles O, Kleuser B, Limosin F, Gulbins E, Kornhuber J, Meunier E, Hoertel N, and Cougoule C
- Subjects
- Animals, Mice, Anti-Inflammatory Agents pharmacology, Anti-Inflammatory Agents therapeutic use, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Ceramides, Disease Models, Animal, Fluoxetine pharmacology, Fluoxetine therapeutic use, SARS-CoV-2, COVID-19 Drug Treatment
- Abstract
The coronavirus disease 2019 (COVID-19) pandemic continues to cause significant morbidity and mortality worldwide. Since a large portion of the world's population is currently unvaccinated or incompletely vaccinated and has limited access to approved treatments against COVID-19, there is an urgent need to continue research on treatment options, especially those at low cost and which are immediately available to patients, particularly in low- and middle-income countries. Prior in vitro and observational studies have shown that fluoxetine, possibly through its inhibitory effect on the acid sphingomyelinase/ceramide system, could be a promising antiviral and anti-inflammatory treatment against COVID-19. In this report, we evaluated the potential antiviral and anti-inflammatory activities of fluoxetine in a K18-hACE2 mouse model of SARS-CoV-2 infection, and against variants of concern in vitro, i.e., SARS-CoV-2 ancestral strain, Alpha B.1.1.7, Gamma P1, Delta B1.617 and Omicron BA.5. Fluoxetine, administrated after SARS-CoV-2 infection, significantly reduced lung tissue viral titres and expression of several inflammatory markers (i.e., IL-6, TNFα, CCL2 and CXCL10). It also inhibited the replication of all variants of concern in vitro. A modulation of the ceramide system in the lung tissues, as reflected by the increase in the ratio HexCer 16:0/Cer 16:0 in fluoxetine-treated mice, may contribute to explain these effects. Our findings demonstrate the antiviral and anti-inflammatory properties of fluoxetine in a K18-hACE2 mouse model of SARS-CoV-2 infection, and its in vitro antiviral activity against variants of concern, establishing fluoxetine as a very promising candidate for the prevention and treatment of SARS-CoV-2 infection and disease pathogenesis.
- Published
- 2022
- Full Text
- View/download PDF
30. Efficacy and Mode of Action of a Direct Inhibitor of Mycobacterium abscessus InhA.
- Author
-
Alcaraz M, Roquet-Banères F, Leon-Icaza SA, Abendroth J, Boudehen YM, Cougoule C, Edwards TE, and Kremer L
- Subjects
- Antitubercular Agents chemistry, Bacterial Proteins metabolism, Catalase metabolism, Catalase pharmacology, Catalase therapeutic use, Humans, Isoniazid chemistry, Isoniazid pharmacology, Mycolic Acids metabolism, NAD metabolism, Mycobacterium Infections, Nontuberculous drug therapy, Mycobacterium Infections, Nontuberculous microbiology, Mycobacterium abscessus genetics, Prodrugs pharmacology
- Abstract
There is an unmet medical need for effective treatments against Mycobacterium abscessus pulmonary infections, to which cystic fibrosis (CF) patients are particularly vulnerable. Recent studies showed that the antitubercular drug isoniazid is inactive against M. abscessus due to the incapacity of the catalase-peroxidase to convert the pro-drug into a reactive metabolite that inhibits the enoyl-ACP reductase InhA. To validate InhA
MAB as a druggable target in M. abscessus , we assayed the activity of NITD-916, a 4-hydroxy-2-pyridone lead candidate initially described as a direct inhibitor of InhA that bypasses KatG bioactivation in Mycobacterium tuberculosis . The compound displayed low MIC values against rough and smooth clinical isolates in vitro and significantly reduced the bacterial burden inside human macrophages. Moreover, treatment with NITD-916 reduced the number and size of intracellular mycobacterial cords, regarded as markers of the severity of the infection. Importantly, NITD-916 significantly lowered the M. abscessus burden in CF-derived lung airway organoids. From a mechanistic perspective, NITD-916 abrogated de novo synthesis of mycolic acids and NITD-916-resistant spontaneous mutants harbored point mutations in InhAMAB at residue 96. That NITD-916 targets InhAMAB directly without activation requirements was confirmed genetically and by resolving the crystal structure of the protein in complex with NADH and NITD-916. These findings collectively indicate that InhAMAB is an attractive target to be exploited for future chemotherapeutic developments against this difficult-to-treat mycobacterium and highlight the potential of NITD-916 derivatives for further evaluation in preclinical settings.- Published
- 2022
- Full Text
- View/download PDF
31. Antidepressant Use and Its Association with 28-Day Mortality in Inpatients with SARS-CoV-2: Support for the FIASMA Model against COVID-19.
- Author
-
Hoertel N, Sánchez-Rico M, Kornhuber J, Gulbins E, Reiersen AM, Lenze EJ, Fritz BA, Jalali F, Mills EJ, Cougoule C, Carpinteiro A, Mühle C, Becker KA, Boulware DR, Blanco C, Alvarado JM, Strub-Wourgaft N, Lemogne C, Limosin F, and On Behalf Of Ap-Hp/Université Paris Cité/Inserm Covid-Research Collaboration Ap-Hp Covid Cdr Initiative And Entrepôt de Données de Santé Ap-Hp Consortium
- Abstract
To reduce Coronavirus Disease 2019 (COVID-19)-related mortality and morbidity, widely available oral COVID-19 treatments are urgently needed. Certain antidepressants, such as fluvoxamine or fluoxetine, may be beneficial against COVID-19. We included 388,945 adult inpatients who tested positive for SARS-CoV-2 at 36 AP−HP (Assistance Publique−Hôpitaux de Paris) hospitals from 2 May 2020 to 2 November 2021. We compared the prevalence of antidepressant use at admission in a 1:1 ratio matched analytic sample with and without COVID-19 (N = 82,586), and assessed its association with 28-day all-cause mortality in a 1:1 ratio matched analytic sample of COVID-19 inpatients with and without antidepressant use at admission (N = 1482). Antidepressant use was significantly less prevalent in inpatients with COVID-19 than in a matched control group of inpatients without COVID-19 (1.9% versus 4.8%; Odds Ratio (OR) = 0.38; 95%CI = 0.35−0.41, p < 0.001). Antidepressant use was significantly associated with reduced 28-day mortality among COVID-19 inpatients (12.8% versus 21.2%; OR = 0.55; 95%CI = 0.41−0.72, p < 0.001), particularly at daily doses of at least 40 mg fluoxetine equivalents. Antidepressants with high FIASMA (Functional Inhibitors of Acid Sphingomyelinase) activity seem to drive both associations. These treatments may reduce SARS-CoV-2 infections and COVID-19-related mortality in inpatients, and may be appropriate for prophylaxis and/or COVID-19 therapy for outpatients or inpatients.
- Published
- 2022
- Full Text
- View/download PDF
32. Caspase-1-driven neutrophil pyroptosis and its role in host susceptibility to Pseudomonas aeruginosa.
- Author
-
Santoni K, Pericat D, Gorse L, Buyck J, Pinilla M, Prouvensier L, Bagayoko S, Hessel A, Leon-Icaza SA, Bellard E, Mazères S, Doz-Deblauwe E, Winter N, Paget C, Girard JP, Pham CTN, Cougoule C, Poincloux R, Lamkanfi M, Lefrançais E, Meunier E, and Planès R
- Subjects
- Animals, Apoptosis Regulatory Proteins genetics, Caspase 1 metabolism, Exotoxins metabolism, Humans, Interleukin-1beta metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Neutrophils microbiology, Pseudomonas aeruginosa metabolism, Inflammasomes metabolism, Pyroptosis
- Abstract
Multiple regulated neutrophil cell death programs contribute to host defense against infections. However, despite expressing all necessary inflammasome components, neutrophils are thought to be generally defective in Caspase-1-dependent pyroptosis. By screening different bacterial species, we found that several Pseudomonas aeruginosa (P. aeruginosa) strains trigger Caspase-1-dependent pyroptosis in human and murine neutrophils. Notably, deletion of Exotoxins U or S in P. aeruginosa enhanced neutrophil death to Caspase-1-dependent pyroptosis, suggesting that these exotoxins interfere with this pathway. Mechanistically, P. aeruginosa Flagellin activates the NLRC4 inflammasome, which supports Caspase-1-driven interleukin (IL)-1β secretion and Gasdermin D (GSDMD)-dependent neutrophil pyroptosis. Furthermore, P. aeruginosa-induced GSDMD activation triggers Calcium-dependent and Peptidyl Arginine Deaminase-4-driven histone citrullination and translocation of neutrophil DNA into the cell cytosol without inducing extracellular Neutrophil Extracellular Traps. Finally, we show that neutrophil Caspase-1 contributes to IL-1β production and susceptibility to pyroptosis-inducing P. aeruginosa strains in vivo. Overall, we demonstrate that neutrophils are not universally resistant for Caspase-1-dependent pyroptosis., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
33. Human NLRP1 is a sensor of pathogenic coronavirus 3CL proteases in lung epithelial cells.
- Author
-
Planès R, Pinilla M, Santoni K, Hessel A, Passemar C, Lay K, Paillette P, Valadão AC, Robinson KS, Bastard P, Lam N, Fadrique R, Rossi I, Pericat D, Bagayoko S, Leon-Icaza SA, Rombouts Y, Perouzel E, Tiraby M, Zhang Q, Cicuta P, Jouanguy E, Neyrolles O, Bryant CE, Floto AR, Goujon C, Lei FZ, Martin-Blondel G, Silva S, Casanova JL, Cougoule C, Reversade B, Marcoux J, Ravet E, and Meunier E
- Subjects
- Caspase 3 metabolism, Humans, Lung metabolism, Lung virology, Peptide Hydrolases genetics, Peptide Hydrolases metabolism, Phosphate-Binding Proteins genetics, Phosphate-Binding Proteins metabolism, Pore Forming Cytotoxic Proteins genetics, Pore Forming Cytotoxic Proteins metabolism, Pyroptosis, COVID-19 genetics, COVID-19 metabolism, COVID-19 virology, Coronavirus 3C Proteases genetics, Coronavirus 3C Proteases metabolism, Epithelial Cells metabolism, Inflammasomes genetics, Inflammasomes metabolism, NLR Proteins genetics, NLR Proteins metabolism, SARS-CoV-2 enzymology, SARS-CoV-2 genetics, SARS-CoV-2 metabolism, SARS-CoV-2 pathogenicity
- Abstract
Inflammation observed in SARS-CoV-2-infected patients suggests that inflammasomes, proinflammatory intracellular complexes, regulate various steps of infection. Lung epithelial cells express inflammasome-forming sensors and constitute the primary entry door of SARS-CoV-2. Here, we describe that the NLRP1 inflammasome detects SARS-CoV-2 infection in human lung epithelial cells. Specifically, human NLRP1 is cleaved at the Q333 site by multiple coronavirus 3CL proteases, which triggers inflammasome assembly and cell death and limits the production of infectious viral particles. Analysis of NLRP1-associated pathways unveils that 3CL proteases also inactivate the pyroptosis executioner Gasdermin D (GSDMD). Subsequently, caspase-3 and GSDME promote alternative cell pyroptosis. Finally, analysis of pyroptosis markers in plasma from COVID-19 patients with characterized severe pneumonia due to autoantibodies against, or inborn errors of, type I interferons (IFNs) highlights GSDME/caspase-3 as potential markers of disease severity. Overall, our findings identify NLRP1 as a sensor of SARS-CoV-2 infection in lung epithelia., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
34. Nigratine as dual inhibitor of necroptosis and ferroptosis regulated cell death.
- Author
-
Delehouzé C, Comte A, Leon-Icaza SA, Cougoule C, Hauteville M, Goekjian P, Bulinski JC, Dimanche-Boitrel MT, Meunier E, Rousselot M, and Bach S
- Subjects
- Apoptosis, Cell Death physiology, Humans, Necroptosis, Necrosis, Receptor-Interacting Protein Serine-Threonine Kinases metabolism, Tumor Necrosis Factor-alpha metabolism, Ferroptosis
- Abstract
Nigratine (also known as 6E11), a flavanone derivative of a plant natural product, was characterized as highly specific non-ATP competitive inhibitor of RIPK1 kinase, one of the key components of necroptotic cell death signaling. We show here that nigratine inhibited both necroptosis (induced by Tumor Necrosis Factor-α) and ferroptosis (induced by the small molecules glutamate, erastin, RSL3 or cumene hydroperoxide) with EC
50 in the µM range. Taken together, our data showed that nigratine is a dual inhibitor of necroptosis and ferroptosis cell death pathways. These findings open potential new therapeutic avenues for treating complex necrosis-related diseases., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
35. Association between FIASMA psychotropic medications and reduced risk of intubation or death in individuals with psychiatric disorders hospitalized for severe COVID-19: an observational multicenter study.
- Author
-
Hoertel N, Sánchez-Rico M, Gulbins E, Kornhuber J, Carpinteiro A, Abellán M, de la Muela P, Vernet R, Beeker N, Neuraz A, Delcuze A, Alvarado JM, Cougoule C, Meneton P, and Limosin F
- Subjects
- Adult, Hospitalization, Humans, Intubation, Intratracheal, Psychotropic Drugs therapeutic use, SARS-CoV-2, COVID-19, Mental Disorders drug therapy
- Abstract
The acid sphingomyelinase (ASM)/ceramide system may provide a useful framework for better understanding SARS-CoV-2 infection and the repurposing of psychotropic medications functionally inhibiting the acid sphingomyelinase/ceramide system (named FIASMA psychotropic medications) against COVID-19. We examined the potential usefulness of FIASMA psychotropic medications in patients with psychiatric disorders hospitalized for severe COVID-19, in an observational multicenter study conducted at Greater Paris University hospitals. Of 545 adult inpatients, 164 (30.1%) received a FIASMA psychotropic medication upon hospital admission for COVID-19. We compared the composite endpoint of intubation or death between patients who received a psychotropic FIASMA medication at baseline and those who did not in time-to-event analyses adjusted for sociodemographic characteristics, psychiatric and other medical comorbidity, and other medications. FIASMA psychotropic medication use at baseline was significantly associated with reduced risk of intubation or death in both crude (HR = 0.42; 95%CI = 0.31-0.57; p < 0.01) and primary inverse probability weighting (IPW) (HR = 0.50; 95%CI = 0.37-0.67; p < 0.01) analyses. This association was not specific to one FIASMA psychotropic class or medication. Patients taking a FIASMA antidepressant at baseline had a significantly reduced risk of intubation or death compared with those taking a non-FIASMA antidepressant at baseline in both crude (HR = 0.57; 95%CI = 0.38-0.86; p < 0.01) and primary IPW (HR = 0.57; 95%CI = 0.37-0.87; p < 0.01) analyses. These associations remained significant in multiple sensitivity analyses. Our results show the potential importance of the ASM/ceramide system framework in COVID-19 and support the continuation of FIASMA psychotropic medications in these patients and the need of large- scale clinical trials evaluating FIASMA medications, and particularly FIASMA antidepressants, against COVID-19., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
36. Mycobacteria-host interactions in human bronchiolar airway organoids.
- Author
-
Iakobachvili N, Leon-Icaza SA, Knoops K, Sachs N, Mazères S, Simeone R, Peixoto A, Bernard C, Murris-Espin M, Mazières J, Cam K, Chalut C, Guilhot C, López-Iglesias C, Ravelli RBG, Neyrolles O, Meunier E, Lugo-Villarino G, Clevers H, Cougoule C, and Peters PJ
- Subjects
- Humans, Macrophages microbiology, Nontuberculous Mycobacteria, Organoids, Mycobacterium abscessus, Mycobacterium tuberculosis, Tuberculosis microbiology
- Abstract
Respiratory infections remain a major global health concern. Tuberculosis is one of the top 10 causes of death worldwide, while infections with Non-Tuberculous Mycobacteria are rising globally. Recent advances in human tissue modeling offer a unique opportunity to grow different human "organs" in vitro, including the human airway, that faithfully recapitulates lung architecture and function. Here, we have explored the potential of human airway organoids (AOs) as a novel system in which to assess the very early steps of mycobacterial infection. We reveal that Mycobacterium tuberculosis (Mtb) and Mycobacterium abscessus (Mabs) mainly reside as extracellular bacteria and infect epithelial cells with very low efficiency. While the AO microenvironment was able to control, but not eliminate Mtb, Mabs thrives. We demonstrate that AOs responded to infection by modulating cytokine, antimicrobial peptide, and mucin gene expression. Given the importance of myeloid cells in mycobacterial infection, we co-cultured infected AOs with human monocyte-derived macrophages and found that these cells interact with the organoid epithelium. We conclude that adult stem cell (ASC)-derived AOs can be used to decipher very early events of mycobacteria infection in human settings thus offering new avenues for fundamental and therapeutic research., (© 2021 The Authors. Molecular Microbiology published by John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
37. Comorbid medical conditions are a key factor to understand the relationship between psychiatric disorders and COVID-19-related mortality: Results from 49,089 COVID-19 inpatients.
- Author
-
Hoertel N, Sánchez-Rico M, Herrera-Morueco JJ, de la Muela P, Gulbins E, Kornhuber J, Carpinteiro A, Becker KA, Cougoule C, and Limosin F
- Subjects
- Comorbidity, Humans, Inpatients psychology, COVID-19, Mental Disorders epidemiology
- Published
- 2022
- Full Text
- View/download PDF
38. Association Between FIASMAs and Reduced Risk of Intubation or Death in Individuals Hospitalized for Severe COVID-19: An Observational Multicenter Study.
- Author
-
Hoertel N, Sánchez-Rico M, Gulbins E, Kornhuber J, Carpinteiro A, Lenze EJ, Reiersen AM, Abellán M, de la Muela P, Vernet R, Blanco C, Cougoule C, Beeker N, Neuraz A, Gorwood P, Alvarado JM, Meneton P, and Limosin F
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, COVID-19 Testing trends, Cohort Studies, Enzyme Inhibitors pharmacology, Enzyme Inhibitors therapeutic use, Female, Humans, Male, Middle Aged, Mortality trends, Retrospective Studies, Sphingomyelin Phosphodiesterase metabolism, Young Adult, COVID-19 Drug Treatment, COVID-19 enzymology, COVID-19 mortality, Hospitalization trends, Intubation, Intratracheal mortality, Intubation, Intratracheal trends, Sphingomyelin Phosphodiesterase antagonists & inhibitors
- Abstract
Several medications commonly used for a number of medical conditions share a property of functional inhibition of acid sphingomyelinase (ASM), or FIASMA. Preclinical and clinical evidence suggest that the ASM/ceramide system may be central to severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) infection. We examined the potential usefulness of FIASMA use among patients hospitalized for severe coronavirus disease 2019 (COVID-19) in an observational multicenter study conducted at Greater Paris University hospitals. Of 2,846 adult patients hospitalized for severe COVID-19, 277 (9.7%) were taking an FIASMA medication at the time of their hospital admission. The primary end point was a composite of intubation and/or death. We compared this end point between patients taking vs. not taking an FIASMA medication in time-to-event analyses adjusted for sociodemographic characteristics and medical comorbidities. The primary analysis was a Cox regression model with inverse probability weighting (IPW). Over a mean follow-up of 9.2 days (SD = 12.5), the primary end point occurred in 104 patients (37.5%) receiving an FIASMA medication, and 1,060 patients (41.4%) who did not. Despite being significantly and substantially associated with older age and greater medical severity, FIASMA medication use was significantly associated with reduced likelihood of intubation or death in both crude (hazard ratio (HR) = 0.71, 95% confidence interval (CI) = 0.58-0.87, P < 0.001) and primary IPW (HR = 0.58, 95%CI = 0.46-0.72, P < 0.001) analyses. This association remained significant in multiple sensitivity analyses and was not specific to one particular FIASMA class or medication. These results show the potential importance of the ASM/ceramide system in COVID-19 and support the continuation of FIASMA medications in these patients. Double-blind controlled randomized clinical trials of these medications for COVID-19 are needed., (© 2021 The Authors. Clinical Pharmacology & Therapeutics published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.)
- Published
- 2021
- Full Text
- View/download PDF
39. Repurposing antidepressants inhibiting the sphingomyelinase acid/ceramide system against COVID-19: current evidence and potential mechanisms.
- Author
-
Hoertel N, Sánchez-Rico M, Cougoule C, Gulbins E, Kornhuber J, Carpinteiro A, Becker KA, Reiersen AM, Lenze EJ, Seftel D, Lemogne C, and Limosin F
- Subjects
- Antidepressive Agents pharmacology, Antidepressive Agents therapeutic use, Ceramides, Humans, Prescriptions, Rimonabant, SARS-CoV-2, COVID-19, Sphingomyelin Phosphodiesterase
- Published
- 2021
- Full Text
- View/download PDF
40. A Pulmonary Lactobacillus murinus Strain Induces Th17 and RORγt + Regulatory T Cells and Reduces Lung Inflammation in Tuberculosis.
- Author
-
Bernard-Raichon L, Colom A, Monard SC, Namouchi A, Cescato M, Garnier H, Leon-Icaza SA, Métais A, Dumas A, Corral D, Ghebrendrias N, Guilloton P, Vérollet C, Hudrisier D, Remot A, Langella P, Thomas M, Cougoule C, Neyrolles O, and Lugo-Villarino G
- Subjects
- Animals, Cells, Cultured, Disease Models, Animal, Humans, Lung microbiology, Lymphocyte Activation, Mice, Mice, Inbred C57BL, Pneumonia, Lactobacillus physiology, Lung immunology, Mycobacterium tuberculosis physiology, Nuclear Receptor Subfamily 1, Group F, Member 3 metabolism, T-Lymphocytes, Regulatory immunology, Th17 Cells immunology, Tuberculosis, Pulmonary immunology
- Abstract
The lungs harbor multiple resident microbial communities, otherwise known as the microbiota. There is an emerging interest in deciphering whether the pulmonary microbiota modulate local immunity, and whether this knowledge could shed light on mechanisms operating in the response to respiratory pathogens. In this study, we investigate the capacity of a pulmonary Lactobacillus strain to modulate the lung T cell compartment and assess its prophylactic potential upon infection with Mycobacterium tuberculosis , the etiological agent of tuberculosis. In naive mice, we report that a Lactobacillus murinus ( Lagilactobacillus murinus ) strain (CNCM I-5314) increases the presence of lung Th17 cells and of a regulatory T cell (Treg) subset known as RORγt
+ Tregs. In particular, intranasal but not intragastric administration of CNCM I-5314 increases the expansion of these lung leukocytes, suggesting a local rather than systemic effect. Resident Th17 and RORγt+ Tregs display an immunosuppressive phenotype that is accentuated by CNCM I-5314. Despite the well-known ability of M. tuberculosis to modulate lung immunity, the immunomodulatory effect by CNCM I-5314 is dominant, as Th17 and RORγt+ Tregs are still highly increased in the lung at 42-d postinfection. Importantly, CNCM I-5314 administration in M. tuberculosis -infected mice results in reduction of pulmonary inflammation, without increasing M. tuberculosis burden. Collectively, our findings provide evidence for an immunomodulatory capacity of CNCM I-5314 at steady state and in a model of chronic inflammation in which it can display a protective role, suggesting that L. murinus strains found in the lung may shape local T cells in mice and, perhaps, in humans., (Copyright © 2021 by The American Association of Immunologists, Inc.)- Published
- 2021
- Full Text
- View/download PDF
41. Host phospholipid peroxidation fuels ExoU-dependent cell necrosis and supports Pseudomonas aeruginosa-driven pathology.
- Author
-
Bagayoko S, Leon-Icaza SA, Pinilla M, Hessel A, Santoni K, Péricat D, Bordignon PJ, Moreau F, Eren E, Boyancé A, Naser E, Lefèvre L, Berrone C, Iakobachvili N, Metais A, Rombouts Y, Lugo-Villarino G, Coste A, Attrée I, Frank DW, Clevers H, Peters PJ, Cougoule C, Planès R, and Meunier E
- Subjects
- Animals, Humans, Mice, Mice, Knockout, Necrosis metabolism, Pseudomonas Infections pathology, Pseudomonas aeruginosa metabolism, Virulence physiology, Bacterial Proteins metabolism, Host-Pathogen Interactions physiology, Lipid Peroxidation physiology, Pseudomonas Infections metabolism, Pseudomonas aeruginosa pathogenicity
- Abstract
Regulated cell necrosis supports immune and anti-infectious strategies of the body; however, dysregulation of these processes drives pathological organ damage. Pseudomonas aeruginosa expresses a phospholipase, ExoU that triggers pathological host cell necrosis through a poorly characterized pathway. Here, we investigated the molecular and cellular mechanisms of ExoU-mediated necrosis. We show that cellular peroxidised phospholipids enhance ExoU phospholipase activity, which drives necrosis of immune and non-immune cells. Conversely, both the endogenous lipid peroxidation regulator GPX4 and the pharmacological inhibition of lipid peroxidation delay ExoU-dependent cell necrosis and improve bacterial elimination in vitro and in vivo. Our findings also pertain to the ExoU-related phospholipase from the bacterial pathogen Burkholderia thailandensis, suggesting that exploitation of peroxidised phospholipids might be a conserved virulence mechanism among various microbial phospholipases. Overall, our results identify an original lipid peroxidation-based virulence mechanism as a strong contributor of microbial phospholipase-driven pathology., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
42. Fatty acid oxidation of alternatively activated macrophages prevents foam cell formation, but Mycobacterium tuberculosis counteracts this process via HIF-1α activation.
- Author
-
Genoula M, Marín Franco JL, Maio M, Dolotowicz B, Ferreyra M, Milillo MA, Mascarau R, Moraña EJ, Palmero D, Matteo M, Fuentes F, López B, Barrionuevo P, Neyrolles O, Cougoule C, Lugo-Villarino G, Vérollet C, Sasiain MDC, and Balboa L
- Subjects
- Animals, Lipid Droplets metabolism, Macrophage Activation physiology, Macrophages metabolism, Male, Mice, Inbred BALB C, Mycobacterium tuberculosis physiology, Tuberculosis microbiology, Foam Cells microbiology, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Lipid Metabolism, Macrophages microbiology, Mycobacterium tuberculosis pathogenicity
- Abstract
The ability of Mycobacterium tuberculosis (Mtb) to persist inside host cells relies on metabolic adaptation, like the accumulation of lipid bodies (LBs) in the so-called foamy macrophages (FM), which are favorable to Mtb. The activation state of macrophages is tightly associated to different metabolic pathways, such as lipid metabolism, but whether differentiation towards FM differs between the macrophage activation profiles remains unclear. Here, we aimed to elucidate whether distinct macrophage activation states exposed to a tuberculosis-associated microenvironment or directly infected with Mtb can form FM. We showed that the triggering of signal transducer and activator of transcription 6 (STAT6) in interleukin (IL)-4-activated human macrophages (M(IL-4)) prevents FM formation induced by pleural effusion from patients with tuberculosis. In these cells, LBs are disrupted by lipolysis, and the released fatty acids enter the β-oxidation (FAO) pathway fueling the generation of ATP in mitochondria. Accordingly, murine alveolar macrophages, which exhibit a predominant FAO metabolism, are less prone to become FM than bone marrow derived-macrophages. Interestingly, direct infection of M(IL-4) macrophages with Mtb results in the establishment of aerobic glycolytic pathway and FM formation, which could be prevented by FAO activation or inhibition of the hypoxia-inducible factor 1-alpha (HIF-1α)-induced glycolytic pathway. In conclusion, our results demonstrate that Mtb has a remarkable capacity to induce FM formation through the rewiring of metabolic pathways in human macrophages, including the STAT6-driven alternatively activated program. This study provides key insights into macrophage metabolism and pathogen subversion strategies., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
43. Tuberculosis-associated IFN-I induces Siglec-1 on tunneling nanotubes and favors HIV-1 spread in macrophages.
- Author
-
Dupont M, Souriant S, Balboa L, Vu Manh TP, Pingris K, Rousset S, Cougoule C, Rombouts Y, Poincloux R, Ben Neji M, Allers C, Kaushal D, Kuroda MJ, Benet S, Martinez-Picado J, Izquierdo-Useros N, Sasiain MDC, Maridonneau-Parini I, Neyrolles O, Vérollet C, and Lugo-Villarino G
- Subjects
- Animals, Cells, Cultured, Coinfection immunology, Female, Gene Expression Profiling, HIV Infections, Humans, Macaca mulatta, Male, Nanotubes, Sialic Acid Binding Ig-like Lectin 1 immunology, HIV-1 pathogenicity, Interferon Type I immunology, Macrophages, Alveolar immunology, Macrophages, Alveolar virology, Sialic Acid Binding Ig-like Lectin 1 genetics, Tuberculosis, Pulmonary immunology
- Abstract
While tuberculosis (TB) is a risk factor in HIV-1-infected individuals, the mechanisms by which Mycobacterium tuberculosis (Mtb) worsens HIV-1 pathogenesis remain scarce. We showed that HIV-1 infection is exacerbated in macrophages exposed to TB-associated microenvironments due to tunneling nanotube (TNT) formation. To identify molecular factors associated with TNT function, we performed a transcriptomic analysis in these macrophages, and revealed the up-regulation of Siglec-1 receptor. Siglec-1 expression depends on Mtb-induced production of type I interferon (IFN-I). In co-infected non-human primates, Siglec-1 is highly expressed by alveolar macrophages, whose abundance correlates with pathology and activation of IFN-I/STAT1 pathway. Siglec-1 localizes mainly on microtubule-containing TNT that are long and carry HIV-1 cargo. Siglec-1 depletion decreases TNT length, diminishes HIV-1 capture and cell-to-cell transfer, and abrogates the exacerbation of HIV-1 infection induced by Mtb. Altogether, we uncover a deleterious role for Siglec-1 in TB-HIV-1 co-infection and open new avenues to understand TNT biology., Competing Interests: MD, SS, LB, TV, KP, SR, CC, YR, RP, MB, CA, DK, MK, SB, JM, NI, MS, IM, ON, CV, GL No competing interests declared, (© 2020, Dupont et al.)
- Published
- 2020
- Full Text
- View/download PDF
44. Editorial: The Mononuclear Phagocyte System in Infectious Disease.
- Author
-
Lugo-Villarino G, Cougoule C, Meunier E, Rombouts Y, Vérollet C, and Balboa L
- Subjects
- HIV Infections immunology, Humans, Mononuclear Phagocyte System, Mycoses immunology, Parasitic Diseases immunology, Phagocytosis immunology, Tuberculosis immunology, Dendritic Cells immunology, Macrophages immunology
- Published
- 2019
- Full Text
- View/download PDF
45. Tuberculosis Exacerbates HIV-1 Infection through IL-10/STAT3-Dependent Tunneling Nanotube Formation in Macrophages.
- Author
-
Souriant S, Balboa L, Dupont M, Pingris K, Kviatcovsky D, Cougoule C, Lastrucci C, Bah A, Gasser R, Poincloux R, Raynaud-Messina B, Al Saati T, Inwentarz S, Poggi S, Moraña EJ, González-Montaner P, Corti M, Lagane B, Vergne I, Allers C, Kaushal D, Kuroda MJ, Sasiain MDC, Neyrolles O, Maridonneau-Parini I, Lugo-Villarino G, and Vérollet C
- Subjects
- Adult, Aged, Animals, Cells, Cultured, Coinfection pathology, Coinfection virology, Female, HIV Infections immunology, HIV Infections pathology, HIV Infections virology, Humans, Macaca mulatta, Macrophage Activation, Macrophages virology, Male, Middle Aged, Mycobacterium tuberculosis, Signal Transduction, Tuberculosis, Pulmonary immunology, Tuberculosis, Pulmonary pathology, Virus Replication, Young Adult, HIV Infections complications, Interleukin-10 metabolism, Macrophages pathology, Nanotubes, STAT3 Transcription Factor metabolism, Tuberculosis, Pulmonary complications
- Abstract
The tuberculosis (TB) bacillus, Mycobacterium tuberculosis (Mtb), and HIV-1 act synergistically; however, the mechanisms by which Mtb exacerbates HIV-1 pathogenesis are not well known. Using in vitro and ex vivo cell culture systems, we show that human M(IL-10) anti-inflammatory macrophages, present in TB-associated microenvironment, produce high levels of HIV-1. In vivo, M(IL-10) macrophages are expanded in lungs of co-infected non-human primates, which correlates with disease severity. Furthermore, HIV-1/Mtb co-infected patients display an accumulation of M(IL-10) macrophage markers (soluble CD163 and MerTK). These M(IL-10) macrophages form direct cell-to-cell bridges, which we identified as tunneling nanotubes (TNTs) involved in viral transfer. TNT formation requires the IL-10/STAT3 signaling pathway, and targeted inhibition of TNTs substantially reduces the enhancement of HIV-1 cell-to-cell transfer and overproduction in M(IL-10) macrophages. Our study reveals that TNTs facilitate viral transfer and amplification, thereby promoting TNT formation as a mechanism to be explored in TB/AIDS potential therapeutics., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
46. Variability in the virulence of specific Mycobacterium tuberculosis clinical isolates alters the capacity of human dendritic cells to signal for T cells.
- Author
-
Ramos-Martinez AG, Valtierra-Alvarado MA, Garcia-Hernandez MH, Hernandez-Pando R, Castañeda-Delgado JE, Cougoule C, Rivas-Santiago B, Neyrolles O, Enciso-Moreno JA, Lugo-Villarino G, and Serrano CJ
- Subjects
- Animals, Humans, Mice, Signal Transduction, Virulence, Dendritic Cells virology, Lymphocyte Activation, Mycobacterium tuberculosis pathogenicity, T-Lymphocytes, Regulatory parasitology
- Abstract
Background: Once in the pulmonary alveoli, Mycobacterium tuberculosis (Mtb) enters into contact with alveolar macrophages and dendritic cells (DCs). DCs represent the link between the innate and adaptive immune system owing to their capacity to be both a sentinel and an orchestrator of the antigen-specific immune responses against Mtb. The effect that the virulence of Mtb has on the interaction between the bacilli and human DCs has not been fully explored., Objective: To evaluate the effect of Mtb virulence on human monocyte-derived DCs., Methods: We exposed human monocyte-derived DCs to Mtb clinical strains (isolated from an epidemiological Mtb diversity study in Mexico) bearing different degrees of virulence and evaluated the capacity of DCs to internalise the bacilli, control intracellular growth, engage cell death pathways, express markers for activation and antigen presentation, and expand to stimulate autologous CD4+ T cells proliferation., Findings: In the case of the hypervirulent Mtb strain (Phenotype 1, strain 9005186, lineage 3), we report that DCs internalise and neutralise intracellular growth of the bacilli, undergo low rates of apoptosis, and contribute poorly to T-cell expansion, as compared to the H37Rv reference strain. In the case of the hypovirulent Mtb strain (Phenotype 4, strain 9985449, lineage 4), although DCs internalise and preclude proliferation of the bacilli, the DCs also display a high level of apoptosis, massive levels of apoptosis that prevent them from maintaining autologous CD4+ T cells in a co-culture system, as compared to H37Rv., Main Conclusions: Our findings suggest that variability in virulence among Mtb clinical strains affects the capacity of DCs to respond to pathogenic challenge and mount an immune response against it, highlighting important parallels to studies previously done in mouse models.
- Published
- 2019
- Full Text
- View/download PDF
47. Effect of the BTK inhibitor ibrutinib on macrophage- and γδ T cell-mediated response against Mycobacterium tuberculosis.
- Author
-
Colado A, Genoula M, Cougoule C, Marín Franco JL, Almejún MB, Risnik D, Kviatcovsky D, Podaza E, Elías EE, Fuentes F, Maridonneau-Parini I, Bezares FR, Fernandez Grecco H, Cabrejo M, Jancic C, Sasiain MDC, Giordano M, Gamberale R, Balboa L, and Borge M
- Subjects
- Adenine analogs & derivatives, Agammaglobulinaemia Tyrosine Kinase antagonists & inhibitors, Humans, Piperidines, Macrophages drug effects, Macrophages metabolism, Mycobacterium tuberculosis drug effects, Protein-Tyrosine Kinases pharmacology, Pyrazoles pharmacology, Pyrimidines pharmacology, Receptors, Antigen, T-Cell, gamma-delta metabolism, T-Lymphocytes drug effects, T-Lymphocytes metabolism
- Published
- 2018
- Full Text
- View/download PDF
48. The C-Type Lectin Receptor DC-SIGN Has an Anti-Inflammatory Role in Human M(IL-4) Macrophages in Response to Mycobacterium tuberculosis .
- Author
-
Lugo-Villarino G, Troegeler A, Balboa L, Lastrucci C, Duval C, Mercier I, Bénard A, Capilla F, Al Saati T, Poincloux R, Kondova I, Verreck FAW, Cougoule C, Maridonneau-Parini I, Sasiain MDC, and Neyrolles O
- Subjects
- Animals, Cell Adhesion Molecules genetics, Cell Survival genetics, Cell Survival immunology, Cytokines metabolism, Female, Gene Expression, Host-Pathogen Interactions genetics, Host-Pathogen Interactions immunology, Humans, Inflammation Mediators metabolism, Lectins, C-Type genetics, Macaca mulatta, Macrophages microbiology, Monocytes immunology, Monocytes metabolism, Phagocytosis immunology, Receptors, Cell Surface genetics, Tuberculosis genetics, Tuberculosis microbiology, Cell Adhesion Molecules metabolism, Lectins, C-Type metabolism, Macrophages immunology, Macrophages metabolism, Mycobacterium tuberculosis immunology, Receptors, Cell Surface metabolism, Tuberculosis immunology, Tuberculosis metabolism
- Abstract
DC-SIGN (CD209/CLEC4L) is a C-type lectin receptor (CLR) that serves as a reliable cell-surface marker of interleukin 4 (IL-4)-activated human macrophages [M(IL-4)], which historically represent the most studied subset within the M2 spectrum of macrophage activation. Although DC-SIGN plays important roles in Mycobacterium tuberculosis (Mtb) interactions with dendritic cells, its contribution to the Mtb-macrophage interaction remains poorly understood. Since high levels of IL-4 are correlated with tuberculosis (TB) susceptibility and progression, we investigated the role of DC-SIGN in M(IL-4) macrophages in the TB context. First, we demonstrate that DC-SIGN expression is present both in CD68
+ macrophages found in tuberculous pulmonary lesions of non-human primates, and in the CD14+ cell population isolated from pleural effusions obtained from TB patients (TB-PE). Likewise, we show that DC-SIGN expression is accentuated in M(IL-4) macrophages derived from peripheral blood CD14+ monocytes isolated from TB patients, or in macrophages stimulated with acellular TB-PE, arguing for the pertinence of DC-SIGN-expressing macrophages in TB. Second, using a siRNA-mediated gene silencing approach, we performed a transcriptomic analysis of DC-SIGN-depleted M(IL-4) macrophages and revealed the upregulation of pro-inflammatory signals in response to challenge with Mtb, as compared to control cells. This pro-inflammatory gene signature was confirmed by RT-qPCR, cytokine/chemokine-based protein array, and ELISA analyses. We also found that inactivation of DC-SIGN renders M(IL-4) macrophages less permissive to Mtb intracellular growth compared to control cells, despite the equal level of bacteria uptake. Last, at the molecular level, we show that DC-SIGN interferes negatively with the pro-inflammatory response and control of Mtb intracellular growth mediated by another CLR, Dectin-1 (CLEC7A). Collectively, this study highlights a dual role for DC-SIGN as, on the one hand, being a host factor granting advantage for Mtb to parasitize macrophages and, on the other hand, representing a molecular switch to turn off the pro-inflammatory response in these cells to prevent potential immunopathology associated to TB.- Published
- 2018
- Full Text
- View/download PDF
49. Podosomes, But Not the Maturation Status, Determine the Protease-Dependent 3D Migration in Human Dendritic Cells.
- Author
-
Cougoule C, Lastrucci C, Guiet R, Mascarau R, Meunier E, Lugo-Villarino G, Neyrolles O, Poincloux R, and Maridonneau-Parini I
- Subjects
- Cell Differentiation, Cells, Cultured, Chemokines immunology, Dendrites immunology, Dendritic Cells enzymology, Humans, Macrophages immunology, Toll-Like Receptors immunology, rho-Associated Kinases immunology, Cell Movement, Dendritic Cells cytology, Endopeptidases metabolism, Podosomes immunology
- Abstract
Dendritic cells (DC) are professional Antigen-Presenting Cells scattered throughout antigen-exposed tissues and draining lymph nodes, and survey the body for pathogens. Their ability to migrate through tissues, a 3D environment, is essential for an effective immune response. Upon infection, recognition of Pathogen-Associated Molecular Patterns (PAMP) by Toll-like receptors (TLR) triggers DC maturation. Mature DC (mDC) essentially use the protease-independent, ROCK-dependent amoeboid mode in vivo , or in collagen matrices in vitro . However, the mechanisms of 3D migration used by human immature DC (iDC) are still poorly characterized. Here, we reveal that human monocyte-derived DC are able to use two migration modes in 3D. In porous matrices of fibrillar collagen I, iDC adopted the amoeboid migration mode. In dense matrices of gelled collagen I or Matrigel, iDC used the protease-dependent, ROCK-independent mesenchymal migration mode. Upon TLR4 activation by LPS, mDC-LPS lose the capacity to form podosomes and degrade the matrix along with impaired mesenchymal migration. TLR2 activation by Pam
3 CSK4 resulted in DC maturation, podosome maintenance, and efficient mesenchymal migration. Under all these conditions, when DC used the mesenchymal mode in dense matrices, they formed 3D podosomes at the tip of cell protrusions. Using PGE2 , known to disrupt podosomes in DC, we observed that the cells remained in an immature status and the mesenchymal migration mode was abolished. We also observed that, while CCL5 (attractant of iDC) enhanced both amoeboid and mesenchymal migration of iDC, CCL19 and CCL21 (attractants of mDC) only enhanced mDC-LPS amoeboid migration without triggering mesenchymal migration. Finally, we examined the migration of iDC in tumor cell spheroids, a tissue-like 3D environment. We observed that iDC infiltrated spheroids of tumor cells using both migration modes. Altogether, these results demonstrate that human DC adopt the mesenchymal mode to migrate in 3D dense environments, which relies on their capacity to form podosomes independent of their maturation status, paving the way of further investigations on in vivo DC migration in dense tissues and its regulation during infections.- Published
- 2018
- Full Text
- View/download PDF
50. The Yin and Yang of Tyrosine Kinase Inhibition During Experimental Polymicrobial Sepsis.
- Author
-
Gonçalves-de-Albuquerque CF, Rohwedder I, Silva AR, Ferreira AS, Kurz ARM, Cougoule C, Klapproth S, Eggersmann T, Silva JD, de Oliveira GP, Capelozzi VL, Schlesinger GG, Costa ER, Estrela Marins RCE, Mócsai A, Maridonneau-Parini I, Walzog B, Macedo Rocco PR, Sperandio M, and de Castro-Faria-Neto HC
- Subjects
- Animals, Cell Adhesion drug effects, Disease Models, Animal, Male, Mice, src-Family Kinases antagonists & inhibitors, Dasatinib pharmacology, Neutrophil Infiltration drug effects, Protein Kinase Inhibitors pharmacology, Sepsis immunology
- Abstract
Neutrophils are the first cells of our immune system to arrive at the site of inflammation. They release cytokines, e.g., chemokines, to attract further immune cells, but also actively start to phagocytose and kill pathogens. In the case of sepsis, this tightly regulated host defense mechanism can become uncontrolled and hyperactive resulting in severe organ damage. Currently, no effective therapy is available to fight sepsis; therefore, novel treatment targets that could prevent excessive inflammatory responses are warranted. Src Family tyrosine Kinases (SFK), a group of tyrosine kinases, have been shown to play a major role in regulating immune cell recruitment and host defense. Leukocytes with SFK depletion display severe spreading and migration defects along with reduced cytokine production. Thus, we investigated the effects of dasatinib, a tyrosine kinase inhibitor, with a strong inhibitory capacity on SFKs during sterile inflammation and polymicrobial sepsis in mice. We found that dasatinib-treated mice displayed diminished leukocyte adhesion and extravasation in tumor necrosis factor-α-stimulated cremaster muscle venules in vivo . In polymicrobial sepsis, sepsis severity, organ damage, and clinical outcome improved in a dose-dependent fashion pointing toward an optimal therapeutic window for dasatinib dosage during polymicrobial sepsis. Dasatinib treatment may, therefore, provide a balanced immune response by preventing an overshooting inflammatory reaction on the one side and bacterial overgrowth on the other side.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.