Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 358
Filtrer
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.
Nat Commun ; 15(1): 6142, 2024 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-39034339

RÉSUMÉ

Myeloid cells are vital components of the immune system and have pivotal functions in orchestrating immune responses. Understanding their functions within the tumor microenvironment and their interactions with tumor-infiltrating lymphocytes presents formidable challenges across diverse cancer types, particularly with regards to cancer immunotherapies. Here, we explore tumor-infiltrating myeloid cells (TIMs) by conducting a pan-cancer analysis using single-cell transcriptomics across eight distinct cancer types, encompassing a total of 192 tumor samples from 129 patients. By examining gene expression patterns and transcriptional activities of TIMs in different cancer types, we discern notable alterations in abundance of TIMs and kinetic behaviors prior to and following immunotherapy. We also identify specific cell-cell interaction targets in immunotherapy; unique and shared regulatory profiles critical for treatment response; and TIMs associated with survival outcomes. Overall, our study illuminates the heterogeneity of TIMs and improves our understanding of tissue-specific and cancer-specific myeloid subsets within the context of tumor immunotherapies.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Cellules myéloïdes , Tumeurs , Analyse sur cellule unique , Microenvironnement tumoral , Humains , Cellules myéloïdes/immunologie , Cellules myéloïdes/métabolisme , Analyse sur cellule unique/méthodes , Tumeurs/immunologie , Tumeurs/génétique , Tumeurs/thérapie , Tumeurs/anatomopathologie , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Immunothérapie/méthodes , Lymphocytes TIL/immunologie , Régulation de l'expression des gènes tumoraux , Transcriptome , Analyse de profil d'expression de gènes
3.
Sci Immunol ; 9(97): eadk3981, 2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39058763

RÉSUMÉ

Tumor-associated macrophages (TAMs) are a heterogeneous population of cells whose phenotypes and functions are shaped by factors that are incompletely understood. Herein, we asked when and where TAMs arise from blood monocytes and how they evolve during tumor development. We initiated pancreatic ductal adenocarcinoma (PDAC) in inducible monocyte fate-mapping mice and combined single-cell transcriptomics and high-dimensional flow cytometry to profile the monocyte-to-TAM transition. We revealed that monocytes differentiate first into a transient intermediate population of TAMs that generates two longer-lived lineages of terminally differentiated TAMs with distinct gene expression profiles, phenotypes, and intratumoral localization. Transcriptome datasets and tumor samples from patients with PDAC evidenced parallel TAM populations in humans and their prognostic associations. These insights will support the design of new therapeutic strategies targeting TAMs in PDAC.


Sujet(s)
Carcinome du canal pancréatique , Monocytes , Tumeurs du pancréas , Macrophages associés aux tumeurs , Animaux , Monocytes/immunologie , Humains , Souris , Macrophages associés aux tumeurs/immunologie , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/anatomopathologie , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Différenciation cellulaire/immunologie , Souris de lignée C57BL , Souris transgéniques
4.
bioRxiv ; 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38979166

RÉSUMÉ

Monocyte-derived macrophages (mo-macs) drive immunosuppression in the tumor microenvironment (TME) and tumor-enhanced myelopoiesis in the bone marrow (BM) fuels these populations. Here, we performed paired transcriptome and chromatin analysis over the continuum of BM myeloid progenitors, circulating monocytes, and tumor-infiltrating mo-macs in mice and in patients with lung cancer to identify myeloid progenitor programs that fuel pro-tumorigenic mo-macs. Analyzing chromatin accessibility and histone mark changes, we show that lung tumors prime accessibility for Nfe2l2 (NRF2) in BM myeloid progenitors as a cytoprotective response to oxidative stress. NRF2 activity is sustained and increased during monocyte differentiation into mo-macs in the lung TME to regulate oxidative stress, in turn promoting metabolic adaptation, resistance to cell death, and contributing to immunosuppressive phenotype. NRF2 genetic deletion and pharmacological inhibition significantly reduced mo-macs' survival and immunosuppression in the TME, enabling NK and T cell therapeutic antitumor immunity and synergizing with checkpoint blockade strategies. Altogether, our study identifies a targetable epigenetic node of myeloid progenitor dysregulation that sustains immunoregulatory mo-macs in the TME.

7.
Nat Commun ; 15(1): 4981, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862483

RÉSUMÉ

Spatial omics data allow in-depth analysis of tissue architectures, opening new opportunities for biological discovery. In particular, imaging techniques offer single-cell resolutions, providing essential insights into cellular organizations and dynamics. Yet, the complexity of such data presents analytical challenges and demands substantial computing resources. Moreover, the proliferation of diverse spatial omics technologies, such as Xenium, MERSCOPE, CosMX in spatial-transcriptomics, and MACSima and PhenoCycler in multiplex imaging, hinders the generality of existing tools. We introduce Sopa ( https://github.com/gustaveroussy/sopa ), a technology-invariant, memory-efficient pipeline with a unified visualizer for all image-based spatial omics. Built upon the universal SpatialData framework, Sopa optimizes tasks like segmentation, transcript/channel aggregation, annotation, and geometric/spatial analysis. Its output includes user-friendly web reports and visualizer files, as well as comprehensive data files for in-depth analysis. Overall, Sopa represents a significant step toward unifying spatial data analysis, enabling a more comprehensive understanding of cellular interactions and tissue organization in biological systems.


Sujet(s)
Logiciel , Humains , Traitement d'image par ordinateur/méthodes , Analyse sur cellule unique/méthodes , Biologie informatique/méthodes , Transcriptome , Animaux
8.
Nat Immunol ; 25(7): 1193-1206, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38834865

RÉSUMÉ

Immune cells experience large cell shape changes during environmental patrolling because of the physical constraints that they encounter while migrating through tissues. These cells can adapt to such deformation events using dedicated shape-sensing pathways. However, how shape sensing affects immune cell function is mostly unknown. Here, we identify a shape-sensing mechanism that increases the expression of the chemokine receptor CCR7 and guides dendritic cell migration from peripheral tissues to lymph nodes at steady state. This mechanism relies on the lipid metabolism enzyme cPLA2, requires nuclear envelope tensioning and is finely tuned by the ARP2/3 actin nucleation complex. We also show that this shape-sensing axis reprograms dendritic cell transcription by activating an IKKß-NF-κB-dependent pathway known to control their tolerogenic potential. These results indicate that cell shape changes experienced by immune cells can define their migratory behavior and immunoregulatory properties and reveal a contribution of the physical properties of tissues to adaptive immunity.


Sujet(s)
Mouvement cellulaire , Cellules dendritiques , Homéostasie , Noeuds lymphatiques , Souris de lignée C57BL , Récepteurs CCR7 , Animaux , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/cytologie , Récepteurs CCR7/métabolisme , Souris , Mouvement cellulaire/immunologie , Forme de la cellule , Facteur de transcription NF-kappa B/métabolisme , Souris knockout , Transduction du signal/immunologie , I-kappa B Kinase/métabolisme , Complexe Arp-2-3/métabolisme
9.
Immunity ; 57(7): 1567-1585.e5, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38821051

RÉSUMÉ

Conventional dendritic cells (cDC) are antigen-presenting cells comprising cDC1 and cDC2, responsible for priming naive CD8+ and CD4+ T cells, respectively. Recent studies have unveiled cDC2 heterogeneity and identified various cDC2 progenitors beyond the common DC progenitor (CDP), hinting at distinct cDC2 lineages. By generating Cd300ciCre-hCD2R26tdTomato reporter mice, we identified a bone marrow pro-cDC2 progenitor exclusively generating cDC2 in vitro and in vivo. Single-cell analyses and multiparametric flow cytometry demonstrated that pro-cDC2 encompasses myeloid-derived pre-cDC2 and lymphoid-derived plasmacytoid DC (pDC)-like precursors differentiating into a transcriptionally convergent cDC2 phenotype. Cd300c-traced cDC2 had distinct transcriptomic profiles, phenotypes, and tissue distributions compared with Ms4a3CreR26tdTomato lineage-traced DC3, a monocyte-DC progenitor (MDP)-derived subset that bypasses CDP. Mice with reduced Cd300c-traced cDC2 showed impaired humoral responses to T cell-dependent antigens. We conclude that progenitors of distinct lineages shape the diversity of mature cDC2 across tissues. Thus, ontogenesis may impact tissue immune responses.


Sujet(s)
Différenciation cellulaire , Lignage cellulaire , Cellules dendritiques , Animaux , Cellules dendritiques/immunologie , Souris , Différenciation cellulaire/immunologie , Souris de lignée C57BL , Analyse sur cellule unique , Cellules souches/cytologie , Cellules souches/immunologie , Cellules souches/métabolisme , Souris transgéniques
10.
Cell Rep ; 43(5): 114250, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38762882

RÉSUMÉ

Acute stroke triggers extensive changes to myeloid immune cell populations in the brain that may be targets for limiting brain damage and enhancing repair. Immunomodulatory approaches will be most effective with precise manipulation of discrete myeloid cell phenotypes in time and space. Here, we investigate how stroke alters mononuclear myeloid cell composition and phenotypes at single-cell resolution and key spatial patterns. Our results show that multiple reactive microglial states and monocyte-derived populations contribute to an extensive myeloid cell repertoire in post-stroke brains. We identify important overlaps and distinctions among different cell types/states that involve ontogeny- and spatial-related properties. Notably, brain connectivity with infarcted tissue underpins the pattern of local and remote altered cell accumulation and reactivity. Our discoveries suggest a global but anatomically governed brain myeloid cell response to stroke that comprises diverse phenotypes arising through intrinsic cell ontogeny factors interacting with exposure to spatially organized brain damage and neuro-axonal cues.


Sujet(s)
Encéphale , Microglie , Cellules myéloïdes , Phénotype , Accident vasculaire cérébral , Animaux , Encéphale/anatomopathologie , Accident vasculaire cérébral/anatomopathologie , Cellules myéloïdes/métabolisme , Microglie/anatomopathologie , Microglie/métabolisme , Mâle , Souris , Souris de lignée C57BL , Monocytes/métabolisme , Monocytes/anatomopathologie
11.
Cancer Cell ; 42(5): 747-758, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38670090

RÉSUMÉ

Cancer is a progressive disease that can develop and evolve over decades, with inflammation playing a central role at each of its stages, from tumor initiation to metastasis. In this context, macrophages represent well-established bridges reciprocally linking inflammation and cancer via an array of diverse functions that have spurred efforts to classify them into subtypes. Here, we discuss the intertwines between macrophages, inflammation, and cancer with an emphasis on temporal dynamics of macrophage diversity and functions in pre-malignancy and cancer. By instilling temporal dynamism into the more static classic view of tumor-associated macrophage biology, we propose a new framework to better contextualize their significance in the inflammatory processes that precede and result from the onset of cancer and shape its evolution.


Sujet(s)
Inflammation , Tumeurs , Microenvironnement tumoral , Macrophages associés aux tumeurs , Animaux , Humains , Inflammation/immunologie , Inflammation/anatomopathologie , Macrophages/immunologie , Tumeurs/immunologie , Tumeurs/anatomopathologie , Microenvironnement tumoral/immunologie , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/métabolisme
12.
J Extracell Vesicles ; 13(3): e12420, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38490958

RÉSUMÉ

High-grade serous carcinoma of the ovary, fallopian tube and peritoneum (HGSC), the most common type of ovarian cancer, ranks among the deadliest malignancies. Many HGSC patients have excess fluid in the peritoneum called ascites. Ascites is a tumour microenvironment (TME) containing various cells, proteins and extracellular vesicles (EVs). We isolated EVs from patients' ascites by orthogonal methods and analyzed them by mass spectrometry. We identified not only a set of 'core ascitic EV-associated proteins' but also defined their subset unique to HGSC ascites. Using single-cell RNA sequencing data, we mapped the origin of HGSC-specific EVs to different types of cells present in ascites. Surprisingly, EVs did not come predominantly from tumour cells but from non-malignant cell types such as macrophages and fibroblasts. Flow cytometry of ascitic cells in combination with analysis of EV protein composition in matched samples showed that analysis of cell type-specific EV markers in HGSC has more substantial prognostic potential than analysis of ascitic cells. To conclude, we provide evidence that proteomic analysis of EVs can define the cellular composition of HGSC TME. This finding opens numerous avenues both for a better understanding of EV's role in tumour promotion/prevention and for improved HGSC diagnostics.


Sujet(s)
Cystadénocarcinome séreux , Vésicules extracellulaires , Tumeurs de l'ovaire , Humains , Femelle , Ascites/métabolisme , Ascites/anatomopathologie , Microenvironnement tumoral , Protéomique , Cystadénocarcinome séreux/diagnostic , Cystadénocarcinome séreux/génétique , Cystadénocarcinome séreux/métabolisme , Vésicules extracellulaires/métabolisme , Tumeurs de l'ovaire/diagnostic
13.
Nat Commun ; 15(1): 2113, 2024 Mar 08.
Article de Anglais | MEDLINE | ID: mdl-38459052

RÉSUMÉ

Macrophages are abundant immune cells in the microenvironment of diffuse large B-cell lymphoma (DLBCL). Macrophage estimation by immunohistochemistry shows varying prognostic significance across studies in DLBCL, and does not provide a comprehensive analysis of macrophage subtypes. Here, using digital spatial profiling with whole transcriptome analysis of CD68+ cells, we characterize macrophages in distinct spatial niches of reactive lymphoid tissues (RLTs) and DLBCL. We reveal transcriptomic differences between macrophages within RLTs (light zone /dark zone, germinal center/ interfollicular), and between disease states (RLTs/ DLBCL), which we then use to generate six spatially-derived macrophage signatures (MacroSigs). We proceed to interrogate these MacroSigs in macrophage and DLBCL single-cell RNA-sequencing datasets, and in gene-expression data from multiple DLBCL cohorts. We show that specific MacroSigs are associated with cell-of-origin subtypes and overall survival in DLBCL. This study provides a spatially-resolved whole-transcriptome atlas of macrophages in reactive and malignant lymphoid tissues, showing biological and clinical significance.


Sujet(s)
Lymphome B diffus à grandes cellules , Humains , Pronostic , Lymphome B diffus à grandes cellules/anatomopathologie , Analyse de profil d'expression de gènes , Transcriptome , Centre germinatif/anatomopathologie , Microenvironnement tumoral/génétique
14.
Cell ; 187(4): 962-980.e19, 2024 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-38309258

RÉSUMÉ

Microglia (MG), the brain-resident macrophages, play major roles in health and disease via a diversity of cellular states. While embryonic MG display a large heterogeneity of cellular distribution and transcriptomic states, their functions remain poorly characterized. Here, we uncovered a role for MG in the maintenance of structural integrity at two fetal cortical boundaries. At these boundaries between structures that grow in distinct directions, embryonic MG accumulate, display a state resembling post-natal axon-tract-associated microglia (ATM) and prevent the progression of microcavities into large cavitary lesions, in part via a mechanism involving the ATM-factor Spp1. MG and Spp1 furthermore contribute to the rapid repair of lesions, collectively highlighting protective functions that preserve the fetal brain from physiological morphogenetic stress and injury. Our study thus highlights key major roles for embryonic MG and Spp1 in maintaining structural integrity during morphogenesis, with major implications for our understanding of MG functions and brain development.


Sujet(s)
Encéphale , Microglie , Axones , Encéphale/cytologie , Encéphale/croissance et développement , Macrophages/physiologie , Microglie/anatomopathologie , Morphogenèse
15.
Immunity ; 57(2): 349-363.e9, 2024 Feb 13.
Article de Anglais | MEDLINE | ID: mdl-38309272

RÉSUMÉ

Microglial reactivity to injury and disease is emerging as a heterogeneous, dynamic, and crucial determinant in neurological disorders. However, the plasticity and fate of disease-associated microglia (DAM) remain largely unknown. We established a lineage tracing system, leveraging the expression dynamics of secreted phosphoprotein 1(Spp1) to label and track DAM-like microglia during brain injury and recovery. Fate mapping of Spp1+ microglia during stroke in juvenile mice revealed an irreversible state of DAM-like microglia that were ultimately eliminated from the injured brain. By contrast, DAM-like microglia in the neonatal stroke models exhibited high plasticity, regaining a homeostatic signature and integrating into the microglial network after recovery. Furthermore, neonatal injury had a lasting impact on microglia, rendering them intrinsically sensitized to subsequent immune challenges. Therefore, our findings highlight the plasticity and innate immune memory of neonatal microglia, shedding light on the fate of DAM-like microglia in various neuropathological conditions.


Sujet(s)
Lésions encéphaliques , Accident vasculaire cérébral , Animaux , Souris , Microglie , Encéphale/métabolisme , Ostéopontine/métabolisme
16.
Science ; 383(6679): eadf6493, 2024 01 12.
Article de Anglais | MEDLINE | ID: mdl-38207030

RÉSUMÉ

Neutrophils are increasingly recognized as key players in the tumor immune response and are associated with poor clinical outcomes. Despite recent advances characterizing the diversity of neutrophil states in cancer, common trajectories and mechanisms governing the ontogeny and relationship between these neutrophil states remain undefined. Here, we demonstrate that immature and mature neutrophils that enter tumors undergo irreversible epigenetic, transcriptional, and proteomic modifications to converge into a distinct, terminally differentiated dcTRAIL-R1+ state. Reprogrammed dcTRAIL-R1+ neutrophils predominantly localize to a glycolytic and hypoxic niche at the tumor core and exert pro-angiogenic function that favors tumor growth. We found similar trajectories in neutrophils across multiple tumor types and in humans, suggesting that targeting this program may provide a means of enhancing certain cancer immunotherapies.


Sujet(s)
Reprogrammation cellulaire , Tumeurs , Néovascularisation pathologique , Granulocytes neutrophiles , Humains , Tumeurs/vascularisation , Tumeurs/immunologie , Granulocytes neutrophiles/immunologie , Protéomique , Reprogrammation cellulaire/génétique , Reprogrammation cellulaire/immunologie , Néovascularisation pathologique/génétique , Néovascularisation pathologique/immunologie , Récepteurs de TRAIL/immunologie , Épigenèse génétique , Hypoxie , Transcription génétique
17.
Sci Rep ; 14(1): 62, 2024 01 02.
Article de Anglais | MEDLINE | ID: mdl-38167979

RÉSUMÉ

The percentage of macrophage subpopulations based on their origins in the adult cochlea remains unclear. This study aimed to elucidate the origins of cochlear macrophages during the onset phase and development of auditory function. We used three types of mice: wildtype ICR mice, colony-stimulating factor 1 receptor (Csf1r)-deficient mice, and Ms4a3Cre-Rosa tdTomato (Ms4a3tdT) transgenic mice. Macrophages were labeled with ionized calcium-binding adapter molecule 1 (Iba1), which is specific to more mature macrophages, and CD11b, which is specific to monocyte lineage. We investigated the spatial and temporal distribution patterns of resident macrophages in the cochlea during the postnatal and early adult stages. During the adult stages, the rate of monocytes recruited from the systemic circulation increased; moreover, Iba1+/CD11b- cochlear macrophages gradually decreased with age. Fate mapping of monocytes using Ms4a3tdT transgenic mice revealed an increased proportion of bone marrow-derived cochlear macrophages in the adult stage. Contrastingly, the proportion of yolk sac- and fetal liver-derived tissue-resident macrophages decreased steadily with age. This heterogeneity could be attributed to differences in environmental niches within the tissue or at the sub-tissue levels. Future studies should investigate the role of cochlear macrophages in homeostasis, inflammation, and other diseases, including infection, autoimmune, and metabolic diseases.


Sujet(s)
Macrophages , Monocytes , Animaux , Souris , Souris de lignée ICR , Macrophages/métabolisme , Souris transgéniques , Cochlée , Homéostasie
18.
J Immunol ; 212(6): 1012-1021, 2024 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-38251913

RÉSUMÉ

It is becoming clear that every organ is seeded by a population of fetal liver-derived macrophages that are replaced at different rates by monocyte-derived macrophages. Using the Ms4a3tdTomato reporter mouse that reports on monocyte-derived alveolar macrophages (Mo-AMs) and our ability to examine AM function using our multichannel intravital microscopy, we examined the fetal-liver derived alveolar macrophage (FL-AM) and Mo-AM populations within the same mouse under various environmental conditions. The experiments unveiled that AMs migrated from alveolus to alveolus and phagocytosed bacteria identically regardless of ontogenic origin. Using 50 PFU of influenza A virus (IAV) determined using the Madin-Darby canine kidney (MDCK) cell line, we noted that both populations were susceptible to IAV-induced immunoparalysis, which also led to impaired phagocytosis of secondary bacterial infections. Both FL-AMs and Mo-AMs were trained by ß-glucan to resist IAV-induced paralysis. Over time (40 wk), Mo-AMs began to outperform FL-AMs, although both populations were still sensitive to IAV. Our data also show that clodronate depletion of AMs leads to replenishment, but by FL-AMs, and these macrophages do show some functional impairment for a limited time. Overall, the system is designed such that new macrophages rapidly assume the function of tissue-resident macrophages when both populations are examined in an identical environment. These data do differ from artificial depletion methods that compare Mo-AMs and FL-AMs.


Sujet(s)
Co-infection , Virus de la grippe A , Animaux , Chiens , Souris , Poumon , Macrophages , Macrophages alvéolaires , Phagocytose , Foie
19.
Nat Cancer ; 5(1): 167-186, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38168935

RÉSUMÉ

Onco-fetal reprogramming of the tumor ecosystem induces fetal developmental signatures in the tumor microenvironment, leading to immunosuppressive features. Here, we employed single-cell RNA sequencing, spatial transcriptomics and bulk RNA sequencing to delineate specific cell subsets involved in hepatocellular carcinoma (HCC) relapse and response to immunotherapy. We identified POSTN+ extracellular matrix cancer-associated fibroblasts (EM CAFs) as a prominent onco-fetal interacting hub, promoting tumor progression. Cell-cell communication and spatial transcriptomics analysis revealed crosstalk and co-localization of onco-fetal cells, including POSTN+ CAFs, FOLR2+ macrophages and PLVAP+ endothelial cells. Further analyses suggest an association between onco-fetal reprogramming and epithelial-mesenchymal transition (EMT), tumor cell proliferation and recruitment of Treg cells, ultimately influencing early relapse and response to immunotherapy. In summary, our study identifies POSTN+ CAFs as part of the HCC onco-fetal niche and highlights its potential influence in EMT, relapse and immunotherapy response, paving the way for the use of onco-fetal signatures for therapeutic stratification.


Sujet(s)
Carcinome hépatocellulaire , Récepteur-2 des folates , Tumeurs du foie , Humains , Carcinome hépatocellulaire/thérapie , Carcinome hépatocellulaire/génétique , Tumeurs du foie/thérapie , Tumeurs du foie/génétique , Écosystème , Cellules endothéliales , Mouvement cellulaire/génétique , Maladie chronique , Récidive , Immunothérapie , Microenvironnement tumoral/génétique
20.
Nat Commun ; 15(1): 811, 2024 Jan 27.
Article de Anglais | MEDLINE | ID: mdl-38280871

RÉSUMÉ

Eosinophils are a group of granulocytes well known for their capacity to protect the host from parasites and regulate immune function. Diverse biological roles for eosinophils have been increasingly identified, but the developmental pattern and regulation of the eosinophil lineage remain largely unknown. Herein, we utilize the zebrafish model to analyze eosinophilic cell differentiation, distribution, and regulation. By identifying eslec as an eosinophil lineage-specific marker, we establish a Tg(eslec:eGFP) reporter line, which specifically labeled cells of the eosinophil lineage from early life through adulthood. Spatial-temporal analysis of eslec+ cells demonstrates their organ distribution from larval stage to adulthood. By single-cell RNA-Seq analysis, we decipher the eosinophil lineage cells from lineage-committed progenitors to mature eosinophils. Through further genetic analysis, we demonstrate the role of Cebp1 in balancing neutrophil and eosinophil lineages, and a Cebp1-Cebpß transcriptional axis that regulates the commitment and differentiation of the eosinophil lineage. Cross-species functional comparisons reveals that zebrafish Cebp1 is the functional orthologue of human C/EBPεP27 in suppressing eosinophilopoiesis. Our study characterizes eosinophil development in multiple dimensions including spatial-temporal patterns, expression profiles, and genetic regulators, providing for a better understanding of eosinophilopoiesis.


Sujet(s)
Protéines liant les séquences stimulatrices de type CCAAT , Granulocytes éosinophiles , Danio zébré , Animaux , Humains , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/métabolisme , Différenciation cellulaire/génétique , Granulocytes éosinophiles/métabolisme , Granulocytes neutrophiles/métabolisme , Danio zébré/génétique , Protéines liant les séquences stimulatrices de type CCAAT/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE