Your browser doesn't support javascript.
loading
TFEB signaling attenuates NLRP3-driven inflammatory responses in severe asthma.
Theofani, Efthymia; Semitekolou, Maria; Samitas, Konstantinos; Mais, Annie; Galani, Ioanna E; Triantafyllia, Vasiliki; Lama, Joanna; Morianos, Ioannis; Stavropoulos, Athanasios; Jeong, Se-Jin; Andreakos, Evangelos; Razani, Babak; Rovina, Nikoletta; Xanthou, Georgina.
Afiliação
  • Theofani E; Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.
  • Semitekolou M; 1st Department of Respiratory Medicine, Medical School, 'Sotiria' Athens Chest Diseases Hospital, National Kapodistrian University of Athens, Athens, Greece.
  • Samitas K; Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.
  • Mais A; Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.
  • Galani IE; 7th Respiratory Clinic and Asthma Center of the 'Sotiria' Athens Chest Hospital, Athens, Greece.
  • Triantafyllia V; Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.
  • Lama J; Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, BRFAA, Athens, Greece.
  • Morianos I; Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, BRFAA, Athens, Greece.
  • Stavropoulos A; Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.
  • Jeong SJ; Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.
  • Andreakos E; Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, BRFAA, Athens, Greece.
  • Razani B; Department of Medicine, Cardiovascular Division, and Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.
  • Rovina N; Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, BRFAA, Athens, Greece.
  • Xanthou G; Department of Medicine, Cardiovascular Division, and Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.
Allergy ; 77(7): 2131-2146, 2022 07.
Article em En | MEDLINE | ID: mdl-35038351
ABSTRACT

BACKGROUND:

NLRP3-driven inflammatory responses by circulating and lung-resident monocytes are critical drivers of asthma pathogenesis. Autophagy restrains NLRP3-induced monocyte activation in asthma models. Yet, the effects of autophagy and its master regulator, transcription factor EB (TFEB), on monocyte responses in human asthma remain unexplored. Here, we investigated whether activation of autophagy and TFEB signaling suppress inflammatory monocyte responses in asthmatic individuals.

METHODS:

Peripheral blood CD14+ monocytes from asthmatic patients (n = 83) and healthy controls (n = 46) were stimulated with LPS/ATP to induce NLRP3 activation with or without the autophagy inducer, rapamycin. ASC specks, caspase-1 activation, IL-1ß and IL-18 levels, mitochondrial function, ROS release, and mTORC1 signaling were examined. Autophagy was evaluated by LC3 puncta formation, p62/SQSTM1 degradation and TFEB activation. In a severe asthma (SA) model, we investigated the role of NLRP3 signaling using Nlrp3-/- mice and/or MCC950 administration, and the effects of TFEB activation using myeloid-specific TFEB-overexpressing mice or administration of the TFEB activator, trehalose.

RESULTS:

We observed increased NLRP3 inflammasome activation, concomitant with impaired autophagy in circulating monocytes that correlated with asthma severity. SA patients also exhibited mitochondrial dysfunction and ROS accumulation. Autophagy failed to inhibit NLRP3-driven monocyte responses, due to defective TFEB activation and excessive mTORC1 signaling. NLRP3 blockade restrained inflammatory cytokine release and linked airway disease. TFEB activation restored impaired autophagy, attenuated NLRP3-driven pulmonary inflammation, and ameliorated SA phenotype.

CONCLUSIONS:

Our studies uncover a crucial role for TFEB-mediated reprogramming of monocyte inflammatory responses, raising the prospect that this pathway can be therapeutically harnessed for the management of SA.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Asma / Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos / Proteína 3 que Contém Domínio de Pirina da Família NLR Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Revista: Allergy Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Grécia

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Asma / Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos / Proteína 3 que Contém Domínio de Pirina da Família NLR Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Revista: Allergy Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Grécia