Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 6 de 6
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Sci Immunol ; 9(98): eado1227, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093958

RÉSUMÉ

The lung is constantly exposed to airborne pathogens and particles that can cause alveolar damage. Hence, appropriate repair responses are essential for gas exchange and life. Here, we deciphered the spatiotemporal trajectory and function of an atypical population of macrophages after lung injury. Post-influenza A virus (IAV) infection, short-lived monocyte-derived Ly6G-expressing macrophages (Ly6G+ Macs) were recruited to the alveoli of lung perilesional areas. Ly6G+ Macs engulfed immune cells, exhibited a high metabolic potential, and clustered with alveolar type 2 epithelial cells (AT2s) in zones of active epithelial regeneration. Ly6G+ Macs were partially dependent on granulocyte-macrophage colony-stimulating factor and interleukin-4 receptor signaling and were essential for AT2-dependent alveolar regeneration. Similar macrophages were recruited in other models of injury and in the airspaces of lungs from patients with suspected pneumonia. This study identifies perilesional alveolar Ly6G+ Macs as a spatially restricted, short-lived macrophage subset promoting epithelial regeneration postinjury, thus representing an attractive therapeutic target for treating lung damage.


Sujet(s)
Antigènes Ly , Lésion pulmonaire , Macrophages alvéolaires , Souris de lignée C57BL , Régénération , Animaux , Antigènes Ly/métabolisme , Antigènes Ly/immunologie , Souris , Régénération/immunologie , Lésion pulmonaire/immunologie , Macrophages alvéolaires/immunologie , Mâle , Humains , Femelle , Infections à Orthomyxoviridae/immunologie , Alvéoles pulmonaires/immunologie , Virus de la grippe A/immunologie , Virus de la grippe A/physiologie
2.
Rev Med Liege ; 79(4): 230-234, 2024 Apr.
Article de Français | MEDLINE | ID: mdl-38602210

RÉSUMÉ

The fields of neutrophil and endothelial cell biology are being deeply revised. While lung marginated neutrophils have been identified decades ago, their roles in the healthy adult lung are still contentious. Furthermore, while it is now clear that the lung constitutes an important immunological niche, the role of lung endothelial cells has been neglected so far. A better understanding of the role of short-lived neutrophils in contributing to lung endothelial cell physiology will improve our understanding of lung endothelial cell fate and heterogeneity under homeostasis and inflammation. Furthermore, it will provide new mechanistic insights on lung marginated neutrophil function and crosstalk with endothelial cells and provide robust foundations for devising therapeutic approaches in which endothelial cell (dys)functions are involved.


Le domaine de la biologie des neutrophiles et des cellules endothéliales est en pleine révision. Si les neutrophiles marginés pulmonaires ont été identifiés il y a plusieurs décennies, leur rôle au niveau du poumon adulte sain reste controversé. De plus, alors qu'il est maintenant reconnu que le poumon constitue une niche immunologique importante, le rôle des cellules endothéliales au niveau de ces niches a, jusqu'à présent, été négligé. Une meilleure compréhension du rôle des neutrophiles marginés dans un poumon sain ainsi que de leur contribution à la physiologie des cellules endothéliales permettrait d'améliorer nos connaissances concernant la biologie et l'hétérogénéité des cellules endothéliales en conditions d'homéostasie et inflammatoires. Enfin, un aperçu mécanistique des relations entre les neutrophiles marginés pulmonaires et les cellules endothéliales constituerait une base solide à l'élaboration de nouvelles stratégies thérapeutiques lors de dysfonctionnements de l'endothélium.


Sujet(s)
Cellules endothéliales , Granulocytes neutrophiles , Humains , Granulocytes neutrophiles/physiologie , Poumon
3.
Am J Respir Cell Mol Biol ; 70(6): 446-456, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38329817

RÉSUMÉ

Lung macrophages constitute a sophisticated surveillance and defense system that contributes to tissue homeostasis and host defense and allows the host to cope with the myriad of insults and antigens to which the lung mucosa is exposed. As opposed to alveolar macrophages, lung interstitial macrophages (IMs) express high levels of Type 2 major histocompatibility complex (MHC-II), a hallmark of antigen-presenting cells. Here, we showed that lung IMs, like dendritic cells, possess the machinery to present soluble antigens in an MHC-II-restricted way. Using ex vivo ovalbumin (OVA)-specific T cell proliferation assays, we found that OVA-pulsed IMs could trigger OVA-specific CD4+ T cell proliferation and Foxp3 expression through MHC-II-, IL-10-, and transforming growth factor ß-dependent mechanisms. Moreover, we showed that IMs efficiently captured locally instilled antigens in vivo, did not migrate to the draining lymph nodes, and enhanced local interactions with CD4+ T cells in a model of OVA-induced allergic asthma. These results support that IMs can present antigens to CD4+ T cells and trigger regulatory T cells, which might attenuate lung immune responses and have functional consequences for lung immunity and T cell-mediated disorders.


Sujet(s)
Présentation d'antigène , Asthme , Facteurs de transcription Forkhead , Poumon , Ovalbumine , Lymphocytes T régulateurs , Animaux , Lymphocytes T régulateurs/immunologie , Facteurs de transcription Forkhead/métabolisme , Facteurs de transcription Forkhead/immunologie , Ovalbumine/immunologie , Poumon/immunologie , Présentation d'antigène/immunologie , Asthme/immunologie , Souris de lignée C57BL , Souris , Prolifération cellulaire , Antigènes d'histocompatibilité de classe II/immunologie , Antigènes d'histocompatibilité de classe II/métabolisme , Antigènes/immunologie , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/immunologie , Interleukine-10/métabolisme , Interleukine-10/immunologie , Macrophages/immunologie , Macrophages/métabolisme , Activation des lymphocytes/immunologie , Macrophages alvéolaires/immunologie , Macrophages alvéolaires/métabolisme , Souris de lignée BALB C
4.
Nat Immunol ; 24(5): 827-840, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36928411

RÉSUMÉ

Resident tissue macrophages (RTMs) are differentiated immune cells that populate distinct niches and exert important tissue-supportive functions. RTM maintenance is thought to rely either on differentiation from monocytes or on RTM self-renewal. Here, we used a mouse model of inducible lung interstitial macrophage (IM) niche depletion and refilling to investigate the development of IMs in vivo. Using time-course single-cell RNA-sequencing analyses, bone marrow chimeras and gene targeting, we found that engrafted Ly6C+ classical monocytes proliferated locally in a Csf1 receptor-dependent manner before differentiating into IMs. The transition from monocyte proliferation toward IM subset specification was controlled by the transcription factor MafB, while c-Maf specifically regulated the identity of the CD206+ IM subset. Our data provide evidence that, in the mononuclear phagocyte system, the ability to proliferate is not merely restricted to myeloid progenitor cells and mature RTMs but is also a tightly regulated capability of monocytes developing into RTMs in vivo.


Sujet(s)
Macrophages , Monocytes , Animaux , Souris , Différenciation cellulaire , Poumon , Prolifération cellulaire , Facteur de transcription MafB/génétique
5.
J Immunother Cancer ; 9(3)2021 03.
Article de Anglais | MEDLINE | ID: mdl-33712445

RÉSUMÉ

BACKGROUND: High-mobility group box 1 (HMGB1) is a multifunctional redox-sensitive protein involved in various intracellular (eg, chromatin remodeling, transcription, autophagy) and extracellular (inflammation, autoimmunity) processes. Regarding its role in cancer development/progression, paradoxical results exist in the literature and it is still unclear whether HMGB1 mainly acts as an oncogene or a tumor suppressor. METHODS: HMGB1 expression was first assessed in tissue specimens (n=359) of invasive breast, lung and cervical cancer and the two distinct staining patterns detected (nuclear vs cytoplasmic) were correlated to the secretion profile of malignant cells, patient outcomes and the presence of infiltrating immune cells within tumor microenvironment. Using several orthotopic, syngeneic mouse models of basal-like breast (4T1, 67NR and EpRas) or non-small cell lung (TC-1) cancer, the efficacy of several HMGB1 inhibitors alone and in combination with immune checkpoint blockade antibodies (anti-PD-1/PD-L1) was then investigated. Isolated from retrieved tumors, 14 immune cell (sub)populations as well as the activation status of antigen-presenting cells were extensively analyzed in each condition. Finally, the redox state of HMGB1 in tumor-extruded fluids and the influence of different forms (oxidized, reduced or disulfide) on both dendritic cell (DC) and plasmacytoid DC (pDC) activation were determined. RESULTS: Associated with an unfavorable prognosis in human patients, we clearly demonstrated that targeting extracellular HMGB1 elicits a profound remodeling of tumor immune microenvironment for efficient cancer therapy. Indeed, without affecting the global number of (CD45+) immune cells, drastic reductions of monocytic/granulocytic myeloid-derived suppressor cells (MDSC) and regulatory T lymphocytes, a higher M1/M2 ratio of macrophages as well as an increased activation of both DC and pDC were continually observed following HMGB1 inhibition. Moreover, blocking HMGB1 improved the efficacy of anti-PD-1 cancer monoimmunotherapy. We also reported that a significant fraction of HMGB1 encountered within cancer microenvironment (interstitial fluids) is oxidized and, in opposite to its reduced isoform, oxidized HMGB1 acts as a tolerogenic signal in a receptor for advanced glycation endproducts-dependent manner. CONCLUSION: Collectively, we present evidence that extracellular HMGB1 blockade may complement first-generation cancer immunotherapies by remobilizing antitumor immune response.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Tumeurs du sein/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Acide glycyrrhizique/pharmacologie , Protéine HMGB1/antagonistes et inhibiteurs , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs du poumon/traitement médicamenteux , Fragments peptidiques/pharmacologie , Protéines S100/pharmacologie , Microenvironnement tumoral/immunologie , Tumeurs du col de l'utérus/traitement médicamenteux , Immunité acquise/effets des médicaments et des substances chimiques , Animaux , Tumeurs du sein/immunologie , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Femelle , Protéine HMGB1/métabolisme , Humains , Tumeurs du poumon/immunologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Macrophages/métabolisme , Mâle , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris nude , Cellules RAW 264.7 , Transduction du signal , Charge tumorale/effets des médicaments et des substances chimiques , Tumeurs du col de l'utérus/immunologie , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/anatomopathologie
6.
Mucosal Immunol ; 13(1): 96-109, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31628426

RÉSUMÉ

Intestinal epithelial cells (IECs) contribute to the regulation of intestinal homeostasis and inflammation through their interactions with the environment and host immune responses. Yet our understanding of IEC-intrinsic regulatory pathways remains incomplete. Here, we identify the guanine nucleotide exchange factor RABGEF1 as a regulator of intestinal homeostasis and innate pathways dependent on IECs. Mice with IEC-specific Rabgef1 deletion (called Rabgef1IEC-KO mice) developed a delayed spontaneous colitis associated with the local upregulation of IEC chemokine expression. In mouse models of colitis based on Interleukin-10 deficiency or dextran sodium sulfate (DSS) exposure, we found that IEC-intrinsic RABGEF1 deficiency exacerbated development of intestinal pathology and dysregulated IEC innate pathways and chemokine expression. Mechanistically, we showed that RABGEF1 deficiency in mouse IECs in vitro was associated with an impairment of early endocytic events, an increased activation of the p38 mitogen-activated protein kinase (MAPK)-dependent pathway, and increased chemokine secretion. Moreover, we provided evidence that the development of spontaneous colitis was dependent on microbiota-derived signals and intrinsic MYD88-dependent pathways in vivo. Our study identifies mouse RABGEF1 as an important regulator of intestinal inflammation, MYD88-dependent IEC-intrinsic signaling, and chemokine production. This suggests that RABGEF1-dependent pathways represent interesting therapeutic targets for inflammatory conditions in the gut.


Sujet(s)
Colite/métabolisme , Facteurs d'échange de nucléotides guanyliques/métabolisme , Muqueuse intestinale/métabolisme , Microbiote/immunologie , Facteur de différenciation myéloïde-88/métabolisme , Animaux , Colite/génétique , Modèles animaux de maladie humaine , Facteurs d'échange de nucléotides guanyliques/génétique , Homéostasie , Humains , Immunité innée , Inflammation , Souris , Souris de lignée C57BL , Souris knockout , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE