Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.536
Filtrer
1.
Front Immunol ; 15: 1382538, 2024.
Article de Anglais | MEDLINE | ID: mdl-39165364

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with an urgent unmet clinical need for new therapies. Using a combination of in vitro assays and in vivo preclinical models we demonstrate that therapeutic inhibition of the IGF signalling axis promotes the accumulation of CD8+ cytotoxic T cells within the tumour microenvironment of PDAC tumours. Mechanistically, we show that IGF blockade promotes macrophage and fibroblast production of the chemokines CXCL9 and CXCL10 to facilitate CD8+ T cell recruitment and trafficking towards the PDAC tumour. Exploring this pathway further, we show that IGF inhibition leads to increased STAT1 transcriptional activity, correlating with a downregulation of the AKT/STAT3 signalling axis, in turn promoting Cxcl9 and Cxcl10 gene transcription. Using patient derived tumour explants, we also demonstrate that our findings translate into the human setting. PDAC tumours are frequently described as "immunologically cold", therefore bolstering CD8+ T cell recruitment to PDAC tumours through IGF inhibition may serve to improve the efficacy of immune checkpoint inhibitors which rely on the presence of CD8+ T cells in tumours.


Sujet(s)
Carcinome du canal pancréatique , Chimiokine CXCL10 , Chimiokine CXCL9 , Macrophages , Tumeurs du pancréas , Microenvironnement tumoral , Chimiokine CXCL9/métabolisme , Humains , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Animaux , Microenvironnement tumoral/immunologie , Chimiokine CXCL10/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Souris , Somatomédines/métabolisme , Lignée cellulaire tumorale , Lymphocytes T cytotoxiques/immunologie , Facteur de transcription STAT-1/métabolisme , Lymphocytes T CD8+/immunologie , Transduction du signal , Fibroblastes/métabolisme , Fibroblastes/immunologie ,
2.
Cancer Cell ; 42(8): 1370-1385.e9, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39137726

RÉSUMÉ

Tertiary lymphoid structures (TLSs) are associated with enhanced immunity in tumors. However, their formation and functions in colorectal cancer liver metastasis (CRLM) remain unclear. Here, we reveal that intra- and peri-tumor mature TLSs (TLS+) are associated with improved clinical outcomes than TLS- tumors. Using single-cell-RNA-sequencing and spatial-enhanced-resolution-omics-sequencing (Stereo-seq), we reveal that TLS+ tumors are enriched with IgG+ plasma cells (PCs), while TLS- tumors are characterized with IgA+ PCs. By generating TLS-associated PC-derived monoclonal antibodies in vitro, we show that TLS-PCs secrete tumor-targeting antibodies. As the proof-of-concept, we demonstrate the anti-tumor activities of TLS-PC-mAb6 antibody in humanized mouse model of colorectal cancer. We identify a fibroblast lineage secreting CCL19 that facilitates lymphocyte trafficking to TLSs. CCL19 treatment promotes TLS neogenesis and prevents tumor growth in mice. Our data uncover the central role of CCL19+ fibroblasts in TLS formation, which in turn generates therapeutic antibodies to restrict CRLM.


Sujet(s)
Chimiokine CCL19 , Tumeurs colorectales , Immunoglobuline G , Tumeurs du foie , Structures lymphoïdes tertiaires , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/immunologie , Tumeurs colorectales/métabolisme , Animaux , Structures lymphoïdes tertiaires/immunologie , Structures lymphoïdes tertiaires/anatomopathologie , Humains , Tumeurs du foie/immunologie , Tumeurs du foie/secondaire , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Souris , Immunoglobuline G/immunologie , Chimiokine CCL19/métabolisme , Chimiokine CCL19/génétique , Fibroblastes/métabolisme , Fibroblastes/immunologie , Anticorps monoclonaux/pharmacologie , Plasmocytes/immunologie , Plasmocytes/métabolisme , Femelle , Lignée cellulaire tumorale
3.
PLoS Pathog ; 20(8): e1012426, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39110744

RÉSUMÉ

Merkel cell polyomavirus (MCPyV) is the causative agent of the majority of Merkel cell carcinomas (MCC). The virus has limited coding capacity, with its early viral proteins, large T (LT) and small T (sT), being multifunctional and contributing to infection and transformation. A fundamental difference in early viral gene expression between infection and MCPyV-driven tumorigenesis is the expression of a truncated LT (LTtr) in the tumor. In contrast, sT is expressed in both conditions and contributes significantly to oncogenesis. Here, we identified novel functions of early viral proteins by performing genome-wide transcriptome and chromatin studies in primary human fibroblasts. Due to current limitations in infection and tumorigenesis models, we mimic these conditions by ectopically expressing sT, LT or LTtr, individually or in combination, at different time points. In addition to its known function in cell cycle and inflammation modulation, we reveal a fundamentally new function of sT. We show that sT regulates the type I interferon (IFN) response downstream of the type I interferon receptor (IFNAR) by interfering with the interferon-stimulated gene factor 3 (ISGF3)-induced interferon-stimulated gene (ISG) response. Expression of sT leads to a reduction in the expression of interferon regulatory factor 9 (IRF9) which is a central component of the ISGF3 complex. We further show that this function of sT is conserved in BKPyV. We provide a first mechanistic understanding of which early viral proteins trigger and control the type I IFN response, which may influence MCPyV infection, persistence and, during MCC progression, regulation of the tumor microenvironment.


Sujet(s)
Carcinome à cellules de Merkel , Échappement immunitaire , Interféron de type I , Polyomavirus des cellules de Merkel , Infections à polyomavirus , Transduction du signal , Infections à virus oncogènes , Humains , Polyomavirus des cellules de Merkel/immunologie , Interféron de type I/métabolisme , Interféron de type I/immunologie , Carcinome à cellules de Merkel/virologie , Carcinome à cellules de Merkel/immunologie , Transduction du signal/immunologie , Infections à polyomavirus/immunologie , Infections à polyomavirus/virologie , Infections à virus oncogènes/immunologie , Infections à virus oncogènes/virologie , Échappement immunitaire/immunologie , Antigènes des virus oncogènes/métabolisme , Antigènes des virus oncogènes/immunologie , Antigènes des virus oncogènes/génétique , Tumeurs cutanées/immunologie , Tumeurs cutanées/virologie , Tumeurs cutanées/métabolisme , Fibroblastes/virologie , Fibroblastes/métabolisme , Fibroblastes/immunologie
4.
Front Immunol ; 15: 1373464, 2024.
Article de Anglais | MEDLINE | ID: mdl-39185406

RÉSUMÉ

Introduction: In systemic sclerosis (SSc), B-cells are activated and present in the skin and lung of patients where they can interact with fibroblasts. The precise impact and mechanisms of the interaction of B-cells and fibroblasts at the tissular level are poorly studied. Objective: We investigated the impact and mechanisms of B-cell/fibroblast interactions in cocultures between B-cells from patients with SSc and 3-dimensional reconstituted healthy skin model including fibroblasts, keratinocytes and extracellular matrix. Methods: The quantification and description of the B-cell infiltration in 3D cocultures were performed using cells imagery strategy and cytometry. The effect of coculture on the transcriptome of B-cells and fibroblasts was studied with bulk and single-cell RNA sequencing approaches. The mechanisms of this interaction were studied by blocking key cytokines like IL-6 and TNF. Results: We showed a significant infiltration of B-cells in the 3D healthy skin model. The amount but not the depth of infiltration was higher with B-cells from SSc patients and with activated B-cells. B-cell infiltrates were mainly composed of naïve and memory cells, whose frequencies differed depending on B-cells origin and activation state: infiltrated B-cells from patients with SSc showed an activated profile and an overexpression of immunoglobulin genes compared to circulating B-cells before infiltration. Our study has shown for the first time that activated B-cells modified the transcriptomic profile of both healthy and SSc fibroblasts, toward a pro-inflammatory (TNF and IL-17 signaling) and interferon profile, with a key role of the TNF pathway. Conclusion: B-cells and 3D skin cocultures allowed the modelization of B-cells infiltration in tissues observed in SSc, uncovering an influence of the underlying disease and the activation state of B-cells. We showed a pro-inflammatory effect on skin fibroblasts and pro-activation effect on infiltrating B-cells during coculture. This reinforces the role of B-cells in SSc and provide potential targets for future therapeutic approach in this disease.


Sujet(s)
Lymphocytes B , Techniques de coculture , Fibroblastes , Sclérodermie systémique , Peau , Humains , Sclérodermie systémique/immunologie , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/métabolisme , Fibroblastes/immunologie , Fibroblastes/métabolisme , Peau/immunologie , Peau/anatomopathologie , Peau/métabolisme , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Femelle , Communication cellulaire/immunologie , Activation des lymphocytes/immunologie , Adulte d'âge moyen , Mâle , Cellules cultivées , Transcriptome , Adulte , Kératinocytes/immunologie , Kératinocytes/métabolisme , Cytokines/métabolisme
5.
Nat Commun ; 15(1): 5949, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009587

RÉSUMÉ

Bullous pemphigoid (BP) is a type 2 inflammation- and immunity-driven skin disease, yet a comprehensive understanding of the immune landscape, particularly immune-stromal crosstalk in BP, remains elusive. Herein, using single-cell RNA sequencing (scRNA-seq) and in vitro functional analyzes, we pinpoint Th2 cells, dendritic cells (DCs), and fibroblasts as crucial cell populations. The IL13-IL13RA1 ligand-receptor pair is identified as the most significant mediator of immune-stromal crosstalk in BP. Notably, fibroblasts and DCs expressing IL13RA1 respond to IL13-secreting Th2 cells, thereby amplifying Th2 cell-mediated cascade responses, which occurs through the specific upregulation of PLA2G2A in fibroblasts and CCL17 in myeloid cells, creating a positive feedback loop integral to immune-stromal crosstalk. Furthermore, PLA2G2A and CCL17 contribute to an increased titer of pathogenic anti-BP180-NC16A autoantibodies in BP patients. Our work provides a comprehensive insight into BP pathogenesis and shows a mechanism governing immune-stromal interactions, providing potential avenues for future therapeutic research.


Sujet(s)
Chimiokine CCL17 , Cellules dendritiques , Fibroblastes , Pemphigoïde bulleuse , Analyse sur cellule unique , Lymphocytes auxiliaires Th2 , Humains , Pemphigoïde bulleuse/immunologie , Pemphigoïde bulleuse/génétique , Analyse sur cellule unique/méthodes , Fibroblastes/métabolisme , Fibroblastes/immunologie , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Chimiokine CCL17/génétique , Chimiokine CCL17/métabolisme , Lymphocytes auxiliaires Th2/immunologie , Autoanticorps/immunologie , Transcriptome , Interleukine-13/métabolisme , Interleukine-13/génétique , Interleukine-13/immunologie , Collagènes non fibrillaires/immunologie , Collagènes non fibrillaires/génétique , Collagènes non fibrillaires/métabolisme , Inflammation/immunologie , Inflammation/génétique , Inflammation/métabolisme , Analyse de profil d'expression de gènes/méthodes , Mâle , Femelle , Autoantigènes/immunologie , Autoantigènes/métabolisme , Autoantigènes/génétique , , Cellules myéloïdes/métabolisme , Cellules myéloïdes/immunologie , Cellules stromales/métabolisme , Cellules stromales/immunologie
6.
Hum Genomics ; 18(1): 80, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39014455

RÉSUMÉ

BACKGROUND: Keloid is a disease characterized by proliferation of fibrous tissue after the healing of skin tissue, which seriously affects the daily life of patients. However, the clinical treatment of keloids still has limitations, that is, it is not effective in controlling keloids, resulting in a high recurrence rate. Thus, it is urgent to identify new signatures to improve the diagnosis and treatment of keloids. METHOD: Bulk RNA seq and scRNA seq data were downloaded from the GEO database. First, we used WGCNA and MEGENA to co-identify keloid/immune-related DEGs. Subsequently, we used three machine learning algorithms (Randomforest, SVM-RFE, and LASSO) to identify hub immune-related genes of keloid (KHIGs) and investigated the heterogeneous expression of KHIGs during fibroblast subpopulation differentiation using scRNA-seq. Finally, we used HE and Masson staining, quantitative reverse transcription-PCR, western blotting, immunohistochemical, and Immunofluorescent assay to investigate the dysregulated expression and the mechanism of retinoic acid in keloids. RESULTS: In the present study, we identified PTGFR, RBP5, and LIF as KHIGs and validated their diagnostic performance. Subsequently, we constructed a novel artificial neural network molecular diagnostic model based on the transcriptome pattern of KHIGs, which is expected to break through the current dilemma faced by molecular diagnosis of keloids in the clinic. Meanwhile, the constructed IG score can also effectively predict keloid risk, which provides a new strategy for keloid prevention. Additionally, we observed that KHIGs were also heterogeneously expressed in the constructed differentiation trajectories of fibroblast subtypes, which may affect the differentiation of fibroblast subtypes and thus lead to dysregulation of the immune microenvironment in keloids. Finally, we found that retinoic acid may treat or alleviate keloids by inhibiting RBP5 to differentiate pro-inflammatory fibroblasts (PIF) to mesenchymal fibroblasts (MF), which further reduces collagen secretion. CONCLUSION: In summary, the present study provides novel immune signatures (PTGFR, RBP5, and LIF) for keloid diagnosis and treatment, and identifies retinoic acid as potential anti-keloid drugs. More importantly, we provide a new perspective for understanding the interactions between different fibroblast subtypes in keloids and the remodeling of their immune microenvironment.


Sujet(s)
Chéloïde , RNA-Seq , Chéloïde/génétique , Chéloïde/diagnostic , Chéloïde/anatomopathologie , Chéloïde/immunologie , Chéloïde/traitement médicamenteux , Humains , Transcriptome/génétique , Analyse de profil d'expression de gènes , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/immunologie , Réseaux de régulation génique , Trétinoïne/pharmacologie , Trétinoïne/usage thérapeutique , Analyse sur cellule unique/méthodes , Différenciation cellulaire/génétique , Analyse de séquence d'ARN/méthodes , Apprentissage machine , Analyse de l'expression du gène de la cellule unique
7.
Int Immunopharmacol ; 138: 112587, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38972211

RÉSUMÉ

There is a growing trend of applying traditional Chinese medicine (TCM) to treat immune diseases. This study reveals the possible mechanism of luteolin, an active ingredient in the core prescription of TCM, in alleviating systemic sclerosis (SSc) inflammation. Bibliometrics was performed to retrieve the core keywords of SSc inflammation. The key inflammatory indicators in the serum samples of 50 SSc patients were detected by ELISA. Data mining was applied for correlation analysis, association rule analysis, and binary logistic regression analysis on the clinical indicators and medication of 50 SSc patients before and after treatment to determine the core prescription. Network pharmacology was used for identifying candidate genes and pathways; molecular docking was conducted to determine the core monomer components of the prescription, providing a basis for subsequent in vitro molecular mechanism research. The effect of luteolin on SSc-human dermal fibroblasts (HDF) viability and inflammatory factors was evaluated by means of ELISA, RT-PCR, and Western blot. The role of TNF in inflammation was explored by using a TNF overexpression vector, NF-κB inhibitor (PKM2), and SSc-HDF. The involvement of TNF/NF-κB pathway was validated by RT-PCR, Western blot, and immunofluorescence. TCM treatment partially corrected the inflammatory changes in SSc patients, indicating its anti-inflammatory effects in the body. Atractylodes, Yam, Astragalus root, Poria cocos, Pinellia ternata, Salvia miltiorrhiza, Safflower, Cassia twig, and Angelica were identified as the core prescriptions for improving inflammatory indicators. Luteolin was the main active ingredient in the prescription and showed a strong binding energy with TNF and NF-κB. Luteolin exerted anti-inflammatory effects in vitro by reducing inflammatory cytokines in SSc-HDF and inhibiting the activation of TNF/NF-κB. Mechanistically, luteolin inhibited the activation of the TNF/NF-κB pathway in SSc-HDF, as manifested by an increase in extranuclear p-P65 and TNF but a decrease in intranuclear p-P65. Interestingly, the addition of PKM2 augmented the therapeutic function of luteolin against inflammation in SSc-HDF. Our study showed the TCM alleviates the inflammatory response of SSc by inhibiting the activation of the TNF/NF-κB pathway and is an effective therapeutic agent for the treatment of SSc.


Sujet(s)
Anti-inflammatoires , Fibroblastes , Lutéoline , Facteur de transcription NF-kappa B , Sclérodermie systémique , Humains , Lutéoline/pharmacologie , Lutéoline/usage thérapeutique , Sclérodermie systémique/traitement médicamenteux , Sclérodermie systémique/immunologie , Facteur de transcription NF-kappa B/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/immunologie , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/pharmacologie , Femelle , Mâle , Biologie des systèmes , Adulte d'âge moyen , Inflammation/traitement médicamenteux , Inflammation/immunologie , Facteur de nécrose tumorale alpha/métabolisme , Simulation de docking moléculaire , Adulte , Transduction du signal/effets des médicaments et des substances chimiques , Cellules cultivées , Médecine traditionnelle chinoise , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Médicaments issus de plantes chinoises/usage thérapeutique , Médicaments issus de plantes chinoises/pharmacologie
8.
J Med Virol ; 96(7): e29811, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39011825

RÉSUMÉ

The recent outbreak of monkeypox virus (MPXV) was unprecedented in its size and distribution. Those living with uncontrolled HIV and low CD4 T cell counts might develop a fulminant clinical mpox course with increased mortality, secondary infections, and necrotizing lesions. Fatal cases display a high and widespread MPXV tissue burden. The underlying pathomechanisms are not fully understood. We report here the pathological findings of an MPXV-driven abscess in gastrocnemius muscle requiring surgery in an immunocompromised patient with severe mpox. Presence of virus particles and infectivity were confirmed by electron microscopy, expansion microscopy, and virus culture, respectively. MPXV tissue distribution by immunohistochemistry (IHC) showed a necrotic core with infection of different cell types. In contrast, at the lesion rim fibroblasts were mainly infected. Immune cells were almost absent in the necrotic core, but were abundant at the infection rim and predominantly macrophages. Further, we detected high amounts of alternatively activated GPNMB+-macrophages at the lesion border. Of note, macrophages only rarely colocalized with virus-infected cells. Insufficient clearance of infected cells and infection of lesion-associated fibroblasts sustained by the abundance of profibrotic macrophages might lead to the coalescing of lesions and the severe and persistent clinical mpox course observed in immunocompromised patients.


Sujet(s)
Sujet immunodéprimé , Virus de la variole simienne , Orthopoxvirose simienne , Muscles squelettiques , Humains , Muscles squelettiques/virologie , Muscles squelettiques/anatomopathologie , Muscles squelettiques/immunologie , Orthopoxvirose simienne/virologie , Orthopoxvirose simienne/immunologie , Virus de la variole simienne/immunologie , Mâle , Macrophages/immunologie , Macrophages/virologie , Fibroblastes/virologie , Fibroblastes/immunologie , Immunohistochimie , Abcès/immunologie , Abcès/virologie , Abcès/anatomopathologie , Adulte d'âge moyen
9.
Respir Res ; 25(1): 273, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997751

RÉSUMÉ

BACKGROUND: Fibroblast differentiation to a myofibroblast phenotype is a feature of airway remodeling in asthma. Lung fibroblasts express the integrin receptor α4ß7 and fibronectin induces myofibroblast differentiation via this receptor. OBJECTIVES: To investigate the role of the ß7 integrin receptor subunit and α4ß7 integrin complex in airway remodeling and airway hyperresponsiveness (AHR) in a murine model of chronic allergen exposure. METHODS: C57BL/6 wild type (WT) and ß7 integrin null mice (ß7 -/-) were sensitized (days 1,10) and challenged with ovalbumin (OVA) three times a week for one or 4 weeks. Similar experiments were performed with WT mice in the presence or absence of α4ß7 blocking antibodies. Bronchoalveolar (BAL) cell counts, AHR, histological evaluation, soluble collagen content, Transforming growth factor-ß (TGFß) and Interleukin-13 (IL13) were measured. Phenotype of fibroblasts cultured from WT and ß7 -/- saline (SAL) and OVA treated mice was evaluated. RESULTS: Eosinophil numbers were similar in WT vs ß7-/- mice. Prolonged OVA exposure in ß7-/- mice was associated with reduced AHR, lung collagen content, peribronchial smooth muscle, lung tissue TGFß and IL13 expression as compared to WT. Similar findings were observed in WT mice treated with α4ß7 blocking antibodies. Fibroblast migration was enhanced in response to OVA in WT but not ß7 -/- fibroblasts. α-SMA and fibronectin expression were reduced in ß7-/- fibroblasts relative to WT. CONCLUSIONS: The ß7 integrin subunit and the α4ß7 integrin complex modulate AHR and airway remodeling in a murine model of allergen exposure. This effect is, at least in part, explained by inhibition of fibroblast activation and is independent of eosinophilic inflammation.


Sujet(s)
Remodelage des voies aériennes , Chaines bêta des intégrines , Souris de lignée C57BL , Souris knockout , Ovalbumine , Animaux , Remodelage des voies aériennes/physiologie , Remodelage des voies aériennes/immunologie , Souris , Ovalbumine/toxicité , Chaines bêta des intégrines/métabolisme , Chaines bêta des intégrines/génétique , Allergènes/immunologie , Allergènes/toxicité , Cellules cultivées , Hyperréactivité bronchique/immunologie , Hyperréactivité bronchique/métabolisme , Hyperréactivité bronchique/physiopathologie , Hyperréactivité bronchique/anatomopathologie , Poumon/métabolisme , Poumon/immunologie , Poumon/anatomopathologie , Modèles animaux de maladie humaine , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/immunologie , Facteur de croissance transformant bêta/métabolisme
10.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-39000078

RÉSUMÉ

The immunogenicity of allogeneic skin fibroblasts in transplantation has been controversial. Whether this controversy comes from a natural heterogeneity among fibroblast subsets or species-specific differences between human and mouse remains to be addressed. In this study, we sought to investigate whether fibroblasts derived from either adult or neonatal human skin tissues could induce different immune responses toward phagocytosis and T cell activation using in vitro co-culture models. Our results indicate that both phagocytosis and T cell proliferation are reduced in the presence of neonatal skin fibroblasts compared to adult skin fibroblasts. We also show that neonatal skin fibroblasts secrete paracrine factors that are responsible for reduced T cell proliferation. In addition, we show that neonatal skin fibroblasts express less class II human leukocyte antigen (HLA) molecules than adult skin fibroblasts after interferon gamma priming, which might also contribute to reduced T cell proliferation. In conclusion, this study supports the use of allogeneic neonatal skin fibroblasts as a readily available cell source for tissue production and transplantation to treat patients with severe injuries.


Sujet(s)
Prolifération cellulaire , Fibroblastes , Peau , Lymphocytes T , Humains , Fibroblastes/métabolisme , Fibroblastes/immunologie , Peau/immunologie , Peau/métabolisme , Peau/cytologie , Nouveau-né , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Activation des lymphocytes/immunologie , Techniques de coculture , Cellules cultivées , Phagocytose , Adulte , Interféron gamma/métabolisme
11.
HLA ; 103(6): e15541, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38923358

RÉSUMÉ

Complications due to HCMV infection or reactivation remain a challenging clinical problem in immunocompromised patients, mainly due to insufficient or absent T-cell functionality. Knowledge of viral targets is crucial to improve monitoring of high-risk patients and optimise antiviral T-cell therapy. To expand the epitope spectrum, genetically-engineered dendritic cells (DCs) and fibroblasts were designed to secrete soluble (s)HLA-A*11:01 and infected with an HCMV mutant lacking immune evasion molecules (US2-6 + 11). More than 700 HLA-A*11:01-restricted epitopes, including more than 50 epitopes derived from a broad range of HCMV open-reading-frames (ORFs) were identified by mass spectrometry and screened for HLA-A*11:01-binding using established prediction tools. The immunogenicity of the 24 highest scoring new candidates was evaluated in vitro in healthy HLA-A*11:01+/HCMV+ donors. Thus, four subdominant epitopes and one immunodominant epitope, derived from the anti-apoptotic protein UL36 and ORFL101C (A11SAL), were identified. Their HLA-A*11:01 complex stability was verified in vitro. In depth analyses revealed highly proliferative and cytotoxic memory T-cell responses against A11SAL, with T-cell responses comparable to the immunodominant HLA-A*02:01-restricted HCMVpp65NLV epitope. A11SAL-specific T cells were also detectable in vivo in immunosuppressed transplant patients and shown to be effective in an in vitro HCMV-infection model, suggesting their crucial role in inhibiting viral replication and improvement of patient's outcome. The developed in vitro pipeline is the first to utilise genetically-engineered DCs to identify naturally presented immunodominant HCMV-derived epitopes. It therefore offers advantages over in silico predictions, is transferable to other HLA alleles, and will significantly expand the repertoire of viral targets to improve therapeutic options.


Sujet(s)
Infections à cytomégalovirus , Cytomegalovirus , Cellules dendritiques , Déterminants antigéniques des lymphocytes T , Épitopes immunodominants , Humains , Cytomegalovirus/immunologie , Infections à cytomégalovirus/immunologie , Épitopes immunodominants/immunologie , Cellules dendritiques/immunologie , Déterminants antigéniques des lymphocytes T/immunologie , Antigène HLA-A11/immunologie , Antigène HLA-A11/génétique , Fibroblastes/immunologie , Fibroblastes/virologie , Cellules présentatrices d'antigène/immunologie
12.
Circ Res ; 134(12): 1703-1717, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38843287

RÉSUMÉ

Fibroblasts are essential for building and maintaining the structural integrity of all organs. Moreover, fibroblasts can acquire an inflammatory phenotype to accommodate immune cells in specific niches and to provide migration, differentiation, and growth factors. In the heart, balancing of fibroblast activity is critical for cardiac homeostasis and optimal organ function during inflammation. Fibroblasts sustain cardiac homeostasis by generating local niche environments that support housekeeping functions and by actively engaging in intercellular cross talk. During inflammatory perturbations, cardiac fibroblasts rapidly switch to an inflammatory state and actively communicate with infiltrating immune cells to orchestrate immune cell migration and activity. Here, we summarize the current knowledge on the molecular landscape of cardiac fibroblasts, focusing on their dual role in promoting tissue homeostasis and modulating immune cell-cardiomyocyte interaction. In addition, we discuss potential future avenues for manipulating cardiac fibroblast activity during myocardial inflammation.


Sujet(s)
Fibroblastes , Homéostasie , Myocarde , Humains , Animaux , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/immunologie , Myocarde/anatomopathologie , Myocarde/immunologie , Myocarde/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/immunologie , Myocardite/immunologie , Myocardite/anatomopathologie , Myocardite/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Communication cellulaire
13.
Front Immunol ; 15: 1385006, 2024.
Article de Anglais | MEDLINE | ID: mdl-38895122

RÉSUMÉ

Osteoarthritis (OA) is the most common form of arthritis, characterized by osteophyte formation, cartilage degradation, and structural and cellular alterations of the synovial membrane. Activated fibroblast-like synoviocytes (FLS) of the synovial membrane have been identified as key drivers, secreting humoral mediators that maintain inflammatory processes, proteases that cause cartilage and bone destruction, and factors that drive fibrotic processes. In normal tissue repair, fibrotic processes are terminated after the damage has been repaired. In fibrosis, tissue remodeling and wound healing are exaggerated and prolonged. Various stressors, including aging, joint instability, and inflammation, lead to structural damage of the joint and micro lesions within the synovial tissue. One result is the reduced production of synovial fluid (lubricants), which reduces the lubricity of the cartilage areas, leading to cartilage damage. In the synovial tissue, a wound-healing cascade is initiated by activating macrophages, Th2 cells, and FLS. The latter can be divided into two major populations. The destructive thymocyte differentiation antigen (THY)1─ phenotype is restricted to the synovial lining layer. In contrast, the THY1+ phenotype of the sublining layer is classified as an invasive one with immune effector function driving synovitis. The exact mechanisms involved in the transition of fibroblasts into a myofibroblast-like phenotype that drives fibrosis remain unclear. The review provides an overview of the phenotypes and spatial distribution of FLS in the synovial membrane of OA, describes the mechanisms of fibroblast into myofibroblast activation, and the metabolic alterations of myofibroblast-like cells.


Sujet(s)
Fibroblastes , Fibrose , Arthrose , Phénotype , Cellules synoviales , Humains , Arthrose/anatomopathologie , Arthrose/immunologie , Arthrose/métabolisme , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/immunologie , Animaux , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Cellules synoviales/immunologie , Membrane synoviale/anatomopathologie , Membrane synoviale/immunologie , Membrane synoviale/métabolisme
14.
J Immunol ; 213(2): 187-203, 2024 07 15.
Article de Anglais | MEDLINE | ID: mdl-38829131

RÉSUMÉ

The RING finger (RNF) family, a group of E3 ubiquitin ligases, plays multiple essential roles in the regulation of innate immunity and resistance to viral infection in mammals. However, it is still unclear whether RNF proteins affect the production of IFN-I and the replication of avian influenza virus (AIV) in ducks. In this article, we found that duck RNF216 (duRNF216) inhibited the duRIG-I signaling pathway. Conversely, duRNF216 deficiency enhanced innate immune responses in duck embryonic fibroblasts. duRNF216 did not interacted with duRIG-I, duMDA5, duMAVS, duSTING, duTBK1, or duIRF7 in the duck RIG-I pathway. However, duRNF216 targeted duTRAF3 and inhibited duMAVS in the recruitment of duTRAF3 in a dose-dependent manner. duRNF216 catalyzed K48-linked polyubiquitination of duck TRAF3, which was degraded by the proteasome pathway. Additionally, AIV PB1 protein competed with duTRAF3 for binding to duRNF216 to reduce degradation of TRAF3 by proteasomes in the cytoplasm, thereby slightly weakening duRNF216-mediated downregulation of IFN-I. Moreover, although duRNF216 downregulated the IFN-ß expression during virus infection, the expression level of IFN-ß in AIV-infected duck embryonic fibroblasts overexpressing duRNF216 was still higher than that in uninfected cells, which would hinder the viral replication. During AIV infection, duRNF216 protein targeted the core protein PB1 of viral polymerase to hinder viral polymerase activity and viral RNA synthesis in the nucleus, ultimately strongly restricting viral replication. Thus, our study reveals a new mechanism by which duRNF216 downregulates innate immunity and inhibits AIV replication in ducks. These findings broaden our understanding of the mechanisms by which the duRNF216 protein affects AIV replication in ducks.


Sujet(s)
Canards , Immunité innée , Sous-type H5N1 du virus de la grippe A , Grippe chez les oiseaux , Transduction du signal , Ubiquitin-protein ligases , Réplication virale , Animaux , Canards/immunologie , Canards/virologie , Réplication virale/immunologie , Transduction du signal/immunologie , Grippe chez les oiseaux/immunologie , Grippe chez les oiseaux/virologie , Sous-type H5N1 du virus de la grippe A/immunologie , Sous-type H5N1 du virus de la grippe A/physiologie , Immunité innée/immunologie , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/immunologie , Fibroblastes/immunologie , Fibroblastes/virologie , Protéines aviaires/immunologie , Protéines aviaires/génétique , Protéines aviaires/métabolisme , Ubiquitination , Protéine-58 à domaine DEAD/métabolisme , Protéine-58 à domaine DEAD/immunologie
15.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 199-205, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38836658

RÉSUMÉ

The present research aimed to conduct a comprehensive critical analysis of existing literature, focusing on the differentiation of myeloid cells from hematopoietic stem cells within the context of immunological tolerance during pregnancy. A comprehensive systematic review was conducted by searching databases including PubMed, Scopus Biomedicine, EBSCOhost, ScienceDirect, Embase, Cochrane Library, and Web of Science. The focus was on the role of myeloid differentiation from hematopoietic stem cells in modulating immune tolerance, particularly during pregnancy and in certain disease states where they act to suppress the immune response. The quality of the evidence gathered was assessed using the GRADE rating system. Our analysis maintains objectivity and independence from the outcomes presented. The current systematic review offers a synthesis of existing research on the transformation of hematopoietic stem cells into fibroblasts across different tissue types. A thorough search of databases such as PubMed, EBSCOhost, Embase, ScienceDirect, Cochrane Library, and Web of Science was performed in conjunction with a specialist in medical information to identify original research on the derivation of fibroblasts following hematopoietic stem cell transplantation. This search yielded a total of 159 studies, of which 10 met the criteria for inclusion in this review. Reflecting on the constraints of this preliminary review, further in-depth and scientific investigations are warranted to comprehensively assess the impact of varied treatments, with a recommendation for clinicians to proceed with increased circumspection. The myeloid differentiation pathway of hematopoietic stem cells is pivotal in modulating the immune environment during pregnancy, supporting the sustenance of a healthy gestational period. Future research in this domain is expected to advance our understanding of the immunological processes occurring at the maternal-fetal boundary.


Sujet(s)
Différenciation cellulaire , Cellules souches hématopoïétiques , Tolérance immunitaire , Femelle , Humains , Cellules souches hématopoïétiques/immunologie , Cellules souches hématopoïétiques/cytologie , Grossesse , Différenciation cellulaire/immunologie , Cellules myéloïdes/immunologie , Cellules myéloïdes/cytologie , Transplantation de cellules souches hématopoïétiques , Fibroblastes/immunologie , Fibroblastes/cytologie
16.
J Virol ; 98(7): e0079124, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38940584

RÉSUMÉ

Fibrocytes were reported to be host cells for HIV-1, but the immunological recognition of HIV-1-infected fibrocytes has not been studied. Here, we investigated the recognition of HIV-1-infected fibrocytes by HIV-1-specific CD8+ T cells. CD8+ T cells specific for five HIV-1 epitopes (HLA-A*24:02-restricted, HLA-B*52:01-restricted, and HLA-C*12:02-restricted epitopes) produced IFN-γ and expressed CD107a after coculture with HIV-1-infected fibrocytes. HIV-1-infected fibrocytes were effectively killed by HIV-1-specific CD8+ T cells. Although it is well known that HIV-1 Nef-mediated downregulation of HLA-A and HLA-B critically affects the T cell recognition of HIV-1-infected CD4+ T cells and HIV-1-infected macrophages, Nef downregulated HLA-A, but not HLA-B, in HIV-1-infected fibrocytes. These findings suggested that HIV-1-specific CD8+ T cells could recognize HIV-1-infected fibrocytes more strongly than HIV-1-infected CD4+ T cells or HIV-1-infected macrophages. HIV-1-infected fibrocytes were also recognized by HIV-1-specific HLA-DR-restricted T cells, indicating that HIV-1-infected fibrocytes can present HIV-1 epitopes to helper T cells. Collectively, these findings suggest that fibrocytes have an important role as antigen-presenting cells during HIV-1 infection. The present study demonstrates effective recognition of HIV-1-infected fibrocytes by HIV-1-specific T cells and suggests possible roles of fibrocytes in the induction and maintenance of HIV-1-specific T cells. IMPORTANCE: Fibrocytes were identified as unique hematopoietic cells with the features of both macrophages and fibroblasts and were demonstrated to be host cells for HIV-1. However, T cell recognition of HIV-1-infected fibrocytes has not been studied. We investigated the recognition of HIV-1-infected fibrocytes by HIV-1-specific T cells. HIV-1-infected fibrocytes were effectively recognized and killed by CD8+ T cells specific for HIV-1 epitopes presented by HLA-A, HLA-B, or HLA-C and were recognized by HIV-1-specific HLA-DR-restricted CD4+ T cells. HIV-1 Nef-mediated downregulation of HLA-A and HLA-B was found in HIV-1-infected CD4+ T cells, whereas Nef did not downregulate HLA-B in HIV-1-infected fibrocytes. These results suggest that HIV-1-specific CD8+ T cells recognize HIV-1-infected fibrocytes more strongly than HIV-1-infected CD4+ T cells. The present study suggests the importance of fibrocytes in the induction and maintenance of HIV-1-specific T cells.


Sujet(s)
Lymphocytes T CD8+ , Régulation négative , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Antigènes HLA-B , Produits du gène nef du virus de l'immunodéficience humaine , Humains , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Produits du gène nef du virus de l'immunodéficience humaine/métabolisme , Produits du gène nef du virus de l'immunodéficience humaine/immunologie , Lymphocytes T CD8+/immunologie , Infections à VIH/immunologie , Infections à VIH/virologie , Antigènes HLA-B/immunologie , Antigènes HLA-B/métabolisme , Fibroblastes/virologie , Fibroblastes/immunologie , Fibroblastes/métabolisme , Lymphocytes T CD4+/immunologie , Déterminants antigéniques des lymphocytes T/immunologie , Macrophages/immunologie , Macrophages/virologie , Macrophages/métabolisme
17.
Front Immunol ; 15: 1337384, 2024.
Article de Anglais | MEDLINE | ID: mdl-38827745

RÉSUMÉ

Fibroblastic reticular cells (FRCs) are a subpopulation of stromal cells modulating the immune environments in health and disease. We have previously shown that activation of TLR9 signaling in FRC in fat-associated lymphoid clusters (FALC) regulate peritoneal immunity via suppressing immune cell recruitment and peritoneal resident macrophage (PRM) retention. However, FRCs are heterogeneous across tissues and organs. The functions of each FRC subset and the regulation of TLR9 in distinct FRC subsets are unknown. Here, we confirmed that specific deletion of TLR9 in FRC improved bacterial clearance and survival during peritoneal infection. Furthermore, using single-cell RNA sequencing, we found two subsets of FRCs (CD55hi and CD55lo) in the mesenteric FALC. The CD55hi FRCs were enriched in gene expression related to extracellular matrix formation. The CD55lo FRCs were enriched in gene expression related to immune response. Interestingly, we found that TLR9 is dominantly expressed in the CD55lo subset. Activation of TLR9 signaling suppressed proliferation, cytokine production, and retinoid metabolism in the CD55lo FRC, but not CD55hi FRC. Notably, we found that adoptive transfer of Tlr9 -/-CD55lo FRC from mesenteric FALC more effectively improved the survival during peritonitis compared with WT-FRC or Tlr9 -/-CD55hi FRC. Furthermore, we identified CD55hi and CD55lo subsets in human adipose tissue-derived FRC and confirmed the suppressive effect of TLR9 on the proliferation and cytokine production in the CD55lo subset. Therefore, inhibition of TLR9 in the CD55lo FRCs from adipose tissue could be a useful strategy to improve the therapeutic efficacy of FRC-based therapy for peritonitis.


Sujet(s)
Fibroblastes , Péritonite , Transduction du signal , Récepteur-9 de type Toll-like , Animaux , Humains , Mâle , Souris , Modèles animaux de maladie humaine , Fibroblastes/métabolisme , Fibroblastes/immunologie , Immunomodulation , Souris de lignée C57BL , Souris knockout , Péritonite/immunologie , Péritonite/métabolisme , Récepteur-9 de type Toll-like/métabolisme , Récepteur-9 de type Toll-like/génétique
18.
Front Immunol ; 15: 1394108, 2024.
Article de Anglais | MEDLINE | ID: mdl-38799455

RÉSUMÉ

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by persistent synovial inflammation and progressive joint destruction. Macrophages are key effector cells that play a central role in RA pathogenesis through their ability to polarize into distinct functional phenotypes. An imbalance favoring pro-inflammatory M1 macrophages over anti-inflammatory M2 macrophages disrupts immune homeostasis and exacerbates joint inflammation. Multiple signaling pathways, including Notch, JAK/STAT, NF-κb, and MAPK, regulate macrophage polarization towards the M1 phenotype in RA. Metabolic reprogramming also contributes to this process, with M1 macrophages prioritizing glycolysis while M2 macrophages utilize oxidative phosphorylation. Redressing this imbalance by modulating macrophage polarization and metabolic state represents a promising therapeutic strategy. Furthermore, complex bidirectional interactions exist between synovial macrophages and fibroblast-like synoviocytes (FLS), forming a self-perpetuating inflammatory loop. Macrophage-derived factors promote aggressive phenotypes in FLS, while FLS-secreted mediators contribute to aberrant macrophage activation. Elucidating the signaling networks governing macrophage polarization, metabolic adaptations, and crosstalk with FLS is crucial to developing targeted therapies that can restore immune homeostasis and mitigate joint pathology in RA.


Sujet(s)
Polyarthrite rhumatoïde , Fibroblastes , Activation des macrophages , Macrophages , Transduction du signal , Membrane synoviale , Humains , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/anatomopathologie , Macrophages/immunologie , Macrophages/métabolisme , Membrane synoviale/métabolisme , Membrane synoviale/immunologie , Membrane synoviale/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/immunologie , Animaux , Activation des macrophages/immunologie , Communication cellulaire/immunologie ,
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE