Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.066
Filtrer
1.
Cell Mol Life Sci ; 81(1): 302, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39008111

RÉSUMÉ

DNAX-activating protein of 12 kDa (DAP12) is a transmembrane adapter protein expressed in lymphoid and myeloid lineage cells. It interacts with several immunoreceptors forming functional complexes that trigger intracellular signaling pathways. One of the DAP12 associated receptors is the triggering receptor expressed on myeloid cells 2 (TREM2). Mutations in both DAP12 and TREM2 have been linked to neurodegenerative diseases. However, mechanisms involved in the regulation of subcellular trafficking and turnover of these proteins are not well understood. Here, we demonstrate that proteasomal degradation of DAP12 is increased in the absence of TREM2. Interestingly, unassembled DAP12 is also retained in early secretory compartments, including the endoplasmic reticulum (ER) and the ER-Golgi intermediate compartment (ERGIC), thereby preventing its transport to the plasma membrane. We also show that unassembled DAP12 interacts with the retention in ER sorting receptor 1 (RER1). The deletion of endogenous RER1 decreases expression of functional TREM2-DAP12 complexes and membrane proximal signaling, and resulted in almost complete inhibition of phagocytic activity in THP-1 differentiated macrophage-like cells. These results indicate that RER1 acts as an important regulator of DAP12 containing immunoreceptor complexes and immune cell function.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Réticulum endoplasmique , Glycoprotéines membranaires , Récepteurs immunologiques , Voie de sécrétion , Humains , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Réticulum endoplasmique/métabolisme , Voie de sécrétion/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Cellules HEK293 , Transduction du signal , Phagocytose/génétique , Macrophages/métabolisme , Transport des protéines , Liaison aux protéines , Animaux , Appareil de Golgi/métabolisme , Protéines du transport vésiculaire/métabolisme , Protéines du transport vésiculaire/génétique , Membrane cellulaire/métabolisme
2.
Life Sci Alliance ; 7(10)2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39025525

RÉSUMÉ

Macrophages have important roles in mammary gland development and tissue homeostasis, but the specific mechanisms that regulate macrophage function need further elucidation. We have identified C/EBPß as an important transcription factor expressed by multiple macrophage populations in the normal mammary gland. Mammary glands from mice with C/EBPß-deficient macrophages (Cebpb ΔM) show a significant decrease in alveolar budding during the diestrus stage of the reproductive cycle, whereas branching morphogenesis remains unchanged. Defects in alveolar budding were found to be the result of both systemic hormones and local macrophage-directed signals. RNA sequencing shows significant changes in PR-responsive genes and alterations in the Wnt landscape of mammary epithelial cells of Cebpb ΔM mice, which regulate stem cell expansion during diestrus. Cebpb ΔM macrophages demonstrate a shift from a pro-inflammatory to a tissue-reparative phenotype, and exhibit increased phagocytic capacity as compared to WT. Finally, Cebpb ΔM macrophages down-regulate Notch2 and Notch3, which normally promote stem cell expansion during alveolar budding. These results suggest that C/EBPß is an important macrophage factor that facilitates macrophage-epithelial crosstalk during a key stage of mammary gland tissue homeostasis.


Sujet(s)
Protéine bêta de liaison aux séquences stimulatrices de type CCAAT , Cycle oestral , Macrophages , Glandes mammaires animales , Animaux , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/métabolisme , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/génétique , Femelle , Souris , Glandes mammaires animales/métabolisme , Glandes mammaires animales/cytologie , Glandes mammaires animales/croissance et développement , Macrophages/métabolisme , Cycle oestral/génétique , Souris knockout , Récepteurs Notch/métabolisme , Récepteurs Notch/génétique , Cellules épithéliales/métabolisme , Phagocytose/génétique , Souris de lignée C57BL , Délétion de gène
3.
Clin Immunol ; 265: 110301, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38944364

RÉSUMÉ

Septic cardiomyopathy (SCM) is characterized by an abnormal inflammatory response and increased mortality. The role of efferocytosis in SCM is not well understood. We used integrated multi-omics analysis to explore the clinical and genetic roles of efferocytosis in SCM. We identified six module genes (ATP11C, CD36, CEBPB, MAPK3, MAPKAPK2, PECAM1) strongly associated with SCM, leading to an accurate predictive model. Subgroups defined by EFFscore exhibited distinct clinical features and immune infiltration levels. Survival analysis showed that the C1 subtype with a lower EFFscore had better survival outcomes. scRNA-seq analysis of peripheral blood mononuclear cells (PBMCs) from sepsis patients identified four genes (CEBPB, CD36, PECAM1, MAPKAPK2) associated with high EFFscores, highlighting their role in SCM. Molecular docking confirmed interactions between diagnostic genes and tamibarotene. Experimental validation supported our computational results. In conclusion, our study identifies a novel efferocytosis-related SCM subtype and diagnostic biomarkers, offering new insights for clinical diagnosis and therapy.


Sujet(s)
Marqueurs biologiques , Cardiomyopathies , Apprentissage machine , Phagocytose , Sepsie , Humains , Cardiomyopathies/génétique , Cardiomyopathies/diagnostic , Pronostic , Mâle , Sepsie/génétique , Sepsie/diagnostic , Phagocytose/génétique , Femelle , Adulte d'âge moyen , Agranulocytes/métabolisme , Sujet âgé , Simulation de docking moléculaire , , Multi-omique
4.
Microb Pathog ; 192: 106681, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38754565

RÉSUMÉ

Tuberculosis (TB) is a major fatal infectious disease globally, exhibiting high morbidity rates and impacting public health and other socio-economic factors. However, some individuals are resistant to TB infection and are referred to as "Resisters". Resisters remain uninfected even after exposure to high load of Mycobacterium tuberculosis (Mtb). To delineate this further, this study aimed to investigate the factors and mechanisms influencing the Mtb resistance phenotype. We assayed the phagocytic capacity of peripheral blood mononuclear cells (PBMCs) collected from Resisters, patients with latent TB infection (LTBI), and patients with active TB (ATB), following infection with fluorescent Mycobacterium bovis Bacillus Calmette-Guérin (BCG). Phagocytosis was stronger in PBMCs from ATB patients, and comparable in LTBI patients and Resisters. Subsequently, phagocytes were isolated and subjected to whole transcriptome sequencing and small RNA sequencing to analyze transcriptional expression profiles and identify potential targets associated with the resistance phenotype. The results revealed that a total of 277 mRNAs, 589 long non-coding RNAs, 523 circular RNAs, and 35 microRNAs were differentially expressed in Resisters and LTBI patients. Further, the endogenous competitive RNA (ceRNA) network was constructed from differentially expressed genes after screening. Bioinformatics, statistical analysis, and quantitative real-time polymerase chain reaction were used for the identification and validation of potential crucial targets in the ceRNA network. As a result, we obtained a ceRNA network that contributes to the resistance phenotype. TCONS_00034796-F3, ENST00000629441-DDX43, hsa-ATAD3A_0003-CYP17A1, and XR_932996.2-CERS1 may be crucial association pairs for resistance to TB infection. Overall, this study demonstrated that the phagocytic capacity of PBMCs was not a determinant of the resistance phenotype and that some non-coding RNAs could be involved in the natural resistance to TB infection through a ceRNA mechanism.


Sujet(s)
Agranulocytes , microARN , Mycobacterium tuberculosis , Phagocytes , Phagocytose , Tuberculose , Humains , Phagocytes/métabolisme , Phagocytes/immunologie , Mycobacterium tuberculosis/génétique , Mycobacterium tuberculosis/immunologie , Tuberculose/génétique , Tuberculose/microbiologie , Tuberculose/immunologie , Phagocytose/génétique , microARN/génétique , microARN/métabolisme , Agranulocytes/immunologie , Agranulocytes/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme , Mâle , Adulte , Analyse de profil d'expression de gènes , Réseaux de régulation génique , Femelle , Transcriptome/génétique , Tuberculose latente/génétique , Tuberculose latente/immunologie , Tuberculose latente/microbiologie , Résistance à la maladie/génétique , ARN messager/génétique , ARN messager/métabolisme , Mycobacterium bovis/immunologie , Adulte d'âge moyen , Biologie informatique/méthodes , Jeune adulte ,
5.
Front Immunol ; 15: 1397541, 2024.
Article de Anglais | MEDLINE | ID: mdl-38774870

RÉSUMÉ

Aim: Despite the significant therapeutic outcomes achieved in systemic treatments for liver hepatocellular carcinoma (LIHC), it is an objective reality that only a low proportion of patients exhibit an improved objective response rate (ORR) to current immunotherapies. Antibody-dependent cellular phagocytosis (ADCP) immunotherapy is considered the new engine for precision immunotherapy. Based on this, we aim to develop an ADCP-based LIHC risk stratification system and screen for relevant targets. Method: Utilizing a combination of single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq data, we screened for ADCP modulating factors in LIHC and identified differentially expressed genes along with their involved functional pathways. A risk scoring model was established by identifying ADCP-related genes with prognostic value through LASSO Cox regression analysis. The risk scoring model was then subjected to evaluations of immune infiltration and immunotherapy relevance, with pan-cancer analysis and in vitro experimental studies conducted on key targets. Results: Building on the research by Kamber RA et al., we identified GYPA, CLDN18, and IRX5 as potential key target genes regulating ADCP in LIHC. These genes demonstrated significant correlations with immune infiltration cells, such as M1-type macrophages, and the effectiveness of immunotherapy in LIHC, as well as a close association with clinical pathological staging and patient prognosis. Pan-cancer analysis revealed that CLDN18 was prognostically and immunologically relevant across multiple types of cancer. Validation through tissue and cell samples confirmed that GYPA and CLDN18 were upregulated in liver cancer tissues and cells. Furthermore, in vitro knockdown of CLDN18 inhibited the malignancy capabilities of liver cancer cells. Conclusion: We have identified an ADCP signature in LIHC comprising three genes. Analysis based on a risk scoring model derived from these three genes, coupled with subsequent experimental validation, confirmed the pivotal role of M1-type macrophages in ADCP within LIHC, establishing CLDN18 as a critical ADCP regulatory target in LIHC.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , RNA-Seq , Humains , Tumeurs du foie/génétique , Tumeurs du foie/immunologie , Tumeurs du foie/thérapie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/thérapie , Pronostic , Immunothérapie/méthodes , Régulation de l'expression des gènes tumoraux , Marqueurs biologiques tumoraux/génétique , Analyse sur cellule unique , Phagocytose/génétique , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Analyse de profil d'expression de gènes , Mâle , Claudines/génétique , Femelle , Analyse de l'expression du gène de la cellule unique
6.
Cell Death Dis ; 15(5): 313, 2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38702326

RÉSUMÉ

CD24 is overexpressed in various tumours and considered a regulator of cell migration, invasion, and proliferation. Recent studies have found that CD24 on ovarian cancer (OC) and triple-negative breast cancer cells interacts with the inhibitory receptor sialic-acid-binding Ig-like lectin 10 (Siglec-10) on tumour-associated macrophages (TAMs) to inhibit phagocytosis by macrophages. Because of its multiple roles in regulating the immune response and tumorigenesis, CD24 is a very promising therapeutic target. However, the regulatory mechanism of CD24 in OC remains unclear. Here, we found that the long noncoding RNA (lncRNA) IL21-AS1, which was upregulated in OC, inhibited macrophage-mediated phagocytosis and promoted OC cell proliferation and apoptosis inhibition. More importantly, after IL21-AS1 knockdown, a significant survival advantage was observed in mice engrafted with tumours. Mechanistically, we identified IL21-AS1 as a hypoxia-induced lncRNA. Moreover, IL21-AS1 increased HIF1α-induced CD24 expression under hypoxic conditions. In parallel, we found that IL21-AS1 acted as a competing endogenous RNA (ceRNA) for miR-561-5p to regulate CD24 expression. Finally, IL21-AS1 increased CD24 expression in OC and facilitated OC progression. Our findings provide a molecular basis for the regulation of CD24, thus highlighting a potential strategy for targeted treatment of OC.


Sujet(s)
Antigènes CD24 , Carcinogenèse , Tumeurs de l'ovaire , Phagocytose , ARN long non codant , Antigènes CD24/métabolisme , Antigènes CD24/génétique , Femelle , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/métabolisme , Phagocytose/génétique , Animaux , Souris , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Lignée cellulaire tumorale , Évolution de la maladie , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux , microARN/métabolisme , microARN/génétique , Souris nude , Apoptose/génétique , Souris de lignée BALB C , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique
7.
Mol Immunol ; 170: 35-45, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38613944

RÉSUMÉ

Microglia play a pivotal role in the pathology of Alzheimer's Disease (AD), with the Triggering Receptor Expressed on Myeloid cells 2 (TREM2) central to their neuroprotective functions. The R47H variant of TREM2 has emerged as a significant genetic risk factor for AD, leading to a loss-of-function phenotype in mouse AD models. This study elucidates the roles of TREM2 in human microglia-like HMC3 cells and the regulation of these functions by SH2-containing inositol-5'-phosphatase 1 (SHIP1). Using stable cell lines expressing wild-type TREM2, the R47H variant, and TREM2-deficient lines, we found that functional TREM2 is essential for the phagocytosis of Aß, lysosomal capacity, and mitochondrial activity. Notably, the R47H variant displayed increased phagocytic activity towards apoptotic neurons. Introducing SHIP1, known to modulate TREM2 signaling in other cells, revealed its role as a negative regulator of these TREM2-mediated functions. Moreover, pharmacological inhibition of both SHIP1 and its isoform SHIP2 amplified Aß phagocytosis and lysosomal capacity, independently of TREM2 or SHIP1 expression, suggesting a potential regulatory role for SHIP2 in these functions. The absence of TREM2, combined with the presence of both SHIP isoforms, suppressed mitochondrial activity. However, pan-SHIP1/2 inhibition enhanced mitochondrial function in these cells. In summary, our findings offer a deeper understanding of the relationship between TREM2 variants and SHIP1 in microglial functions, and emphasize the therapeutic potential of targeting the TREM2 and SHIP1 pathways in microglia for neurodegenerative diseases.


Sujet(s)
Glycoprotéines membranaires , Microglie , Phagocytose , Phosphatidylinositol-3,4,5-trisphosphate 5-phosphatases , Récepteurs immunologiques , Animaux , Humains , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/génétique , Peptides bêta-amyloïdes/métabolisme , Apoptose/génétique , Lignée cellulaire , Lysosomes/métabolisme , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Microglie/métabolisme , Mitochondries/métabolisme , Phagocytose/génétique , Phosphatidylinositol-3,4,5-trisphosphate 5-phosphatases/génétique , Phosphatidylinositol-3,4,5-trisphosphate 5-phosphatases/métabolisme , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Transduction du signal
8.
Biochem Biophys Res Commun ; 715: 150007, 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38678783

RÉSUMÉ

Smad4, a critical mediator of TGF-ß signaling, plays a pivotal role in regulating various cellular functions, including immune responses. In this study, we investigated the impact of Smad4 knockout specifically in macrophages on anti-tumor immunity, focusing on lung metastasis of B16 melanoma cells. Using a mouse model with Smad4 knockout in macrophages established via Lyz2-cre mice and Smad4 flox/flox mice, we demonstrated a significant inhibition of B16 metastasis in the lungs. Interestingly, the inhibition of tumor growth was found to be independent of adaptive immunity, as no significant changes were observed in the numbers or activities of T cells, B cells, or NK cells. Instead, Smad4 knockout led to the emergence of an MCHIIlow CD206high subset of lung interstitial macrophages, characterized by enhanced phagocytosis function. Our findings highlight the crucial role of Smad4 in modulating the innate immune response against tumors and provide insights into potential therapeutic strategies targeting lung interstitial macrophages to enhance anti-tumor immunity.


Sujet(s)
Tumeurs du poumon , Mélanome expérimental , Phagocytose , Protéine Smad-4 , Animaux , Souris , Lignée cellulaire tumorale , Poumon/anatomopathologie , Poumon/immunologie , Poumon/métabolisme , Tumeurs du poumon/secondaire , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Macrophages/immunologie , Macrophages/métabolisme , Macrophages alvéolaires/métabolisme , Macrophages alvéolaires/immunologie , Macrophages alvéolaires/anatomopathologie , Mélanome expérimental/anatomopathologie , Mélanome expérimental/immunologie , Souris de lignée C57BL , Souris knockout , Phagocytose/génétique , Protéine Smad-4/déficit , Protéine Smad-4/génétique , Protéine Smad-4/métabolisme
9.
Aging (Albany NY) ; 16(8): 6883-6897, 2024 04 10.
Article de Anglais | MEDLINE | ID: mdl-38613800

RÉSUMÉ

BACKGROUND: Diabetic foot ulcers (DFUs) pose a serious long-term threat because of elevated mortality and disability risks. Research on its biomarkers is still, however, very limited. In this paper, we have effectively identified biomarkers linked with macrophage excretion in diabetic foot ulcers through the application of bioinformatics and machine learning methodologies. These findings were subsequently validated using external datasets and animal experiments. Such discoveries are anticipated to offer novel insights and approaches for the early diagnosis and treatment of DFU. METHODS: In this work, we used the Gene Expression Omnibus (GEO) database's datasets GSE68183 and GSE80178 as the training dataset to build a gene model using machine learning methods. After that, we used the training and validation sets to validate the model (GSE134431). On the model genes, we performed enrichment analysis using both gene set variant analysis (GSVA) and gene set enrichment analysis (GSEA). Additionally, the model genes were subjected to immunological association and immune function analyses. RESULTS: In this study, PROS1 was identified as a potential key target associated with macrophage efflux in DFU by machine learning and bioinformatics approaches. Subsequently, the key biomarker status of PROS1 in DFU was also confirmed by external datasets. In addition, PROS1 also plays a key role in macrophage exudation in DFU. This gene may be associated with macrophage M1, CD4 memory T cells, naïve B cells, and macrophage M2, and affects IL-17, Rap1, hedgehog, and JAK-STAT signaling pathways. CONCLUSIONS: PROS1 was identified and validated as a biomarker for DFU. This finding has the potential to provide a target for macrophage clearance of DFU.


Sujet(s)
Pied diabétique , Apprentissage machine , Macrophages , Pied diabétique/génétique , Pied diabétique/métabolisme , Macrophages/métabolisme , Animaux , Humains , Phagocytose/génétique , Marqueurs biologiques/métabolisme , Biologie informatique , Souris ,
10.
Front Immunol ; 15: 1355315, 2024.
Article de Anglais | MEDLINE | ID: mdl-38558807

RÉSUMÉ

Macrophage activation syndrome (MAS) is a life-threatening complication of systemic juvenile arthritis, accompanied by cytokine storm and hemophagocytosis. In addition, COVID-19-related hyperinflammation shares clinical features of MAS. Mechanisms that activate macrophages in MAS remain unclear. Here, we identify the role of miRNA in increased phagocytosis and interleukin-12 (IL-12) production by macrophages in a murine model of MAS. MAS significantly increased F4/80+ macrophages and phagocytosis in the mouse liver. Gene expression profile revealed the induction of Fcγ receptor-mediated phagocytosis (FGRP) and IL-12 production in the liver. Phagocytosis pathways such as High-affinity IgE receptor is known as Fc epsilon RI -signaling and pattern recognition receptors involved in the recognition of bacteria and viruses and phagosome formation were also significantly upregulated. In MAS, miR-136-5p and miR-501-3p targeted and caused increased expression of Fcgr3, Fcgr4, and Fcgr1 genes in FGRP pathway and consequent increase in phagocytosis by macrophages, whereas miR-129-1-3p and miR-150-3p targeted and induced Il-12. Transcriptome analysis of patients with MAS revealed the upregulation of FGRP and FCGR gene expression. A target analysis of gene expression data from a patient with MAS discovered that miR-136-5p targets FCGR2A and FCGR3A/3B, the human orthologs of mouse Fcgr3 and Fcgr4, and miR-501-3p targets FCGR1A, the human ortholog of mouse Fcgr1. Together, we demonstrate the novel role of miRNAs during MAS pathogenesis, thereby suggesting miRNA mimic-based therapy to control the hyperactivation of macrophages in patients with MAS as well as use overexpression of FCGR genes as a marker for MAS classification.


Sujet(s)
Syndrome d'activation macrophagique , microARN , Humains , Animaux , Souris , microARN/génétique , microARN/métabolisme , Récepteurs du fragment Fc des IgG/génétique , Syndrome d'activation macrophagique/génétique , Phagocytose/génétique , Interleukine-12
11.
Cell Rep ; 43(4): 114073, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38578825

RÉSUMÉ

Macrophages are central innate immune cells whose function declines with age. The molecular mechanisms underlying age-related changes remain poorly understood, particularly in human macrophages. We report a substantial reduction in phagocytosis, migration, and chemotaxis in human monocyte-derived macrophages (MDMs) from older (>50 years old) compared with younger (18-30 years old) donors, alongside downregulation of transcription factors MYC and USF1. In MDMs from young donors, knockdown of MYC or USF1 decreases phagocytosis and chemotaxis and alters the expression of associated genes, alongside adhesion and extracellular matrix remodeling. A concordant dysregulation of MYC and USF1 target genes is also seen in MDMs from older donors. Furthermore, older age and loss of either MYC or USF1 in MDMs leads to an increased cell size, altered morphology, and reduced actin content. Together, these results define MYC and USF1 as key drivers of MDM age-related functional decline and identify downstream targets to improve macrophage function in aging.


Sujet(s)
Vieillissement , Macrophages , Phagocytose , Protéines proto-oncogènes c-myc , Facteurs de transcription USF , Humains , Macrophages/métabolisme , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Adulte , Facteurs de transcription USF/métabolisme , Facteurs de transcription USF/génétique , Adulte d'âge moyen , Adolescent , Phagocytose/génétique , Jeune adulte , Transcription génétique , Sujet âgé , Chimiotaxie/génétique
12.
Alzheimers Dement ; 20(7): 4914-4934, 2024 07.
Article de Anglais | MEDLINE | ID: mdl-38506634

RÉSUMÉ

BACKGROUND: Variants in ABCA7, a member of the ABC transporter superfamily, have been associated with increased risk for developing late onset Alzheimer's disease (LOAD). METHODS: CRISPR-Cas9 was used to generate an Abca7V1613M variant in mice, modeling the homologous human ABCA7V1599M variant, and extensive characterization was performed. RESULTS: Abca7V1613M microglia show differential gene expression profiles upon lipopolysaccharide challenge and increased phagocytic capacity. Homozygous Abca7V1613M mice display elevated circulating cholesterol and altered brain lipid composition. When crossed with 5xFAD mice, homozygous Abca7V1613M mice display fewer Thioflavin S-positive plaques, decreased amyloid beta (Aß) peptides, and altered amyloid precursor protein processing and trafficking. They also exhibit reduced Aß-associated inflammation, gliosis, and neuronal damage. DISCUSSION: Overall, homozygosity for the Abca7V1613M variant influences phagocytosis, response to inflammation, lipid metabolism, Aß pathology, and neuronal damage in mice. This variant may confer a gain of function and offer a protective effect against Alzheimer's disease-related pathology. HIGHLIGHTS: ABCA7 recognized as a top 10 risk gene for developing Alzheimer's disease. Loss of function mutations result in increased risk for LOAD. V1613M variant reduces amyloid beta plaque burden in 5xFAD mice. V1613M variant modulates APP processing and trafficking in 5xFAD mice. V1613M variant reduces amyloid beta-associated damage in 5xFAD mice.


Sujet(s)
Transporteurs ABC , Maladie d'Alzheimer , Peptides bêta-amyloïdes , Souris transgéniques , Plaque amyloïde , Animaux , Souris , Transporteurs ABC/génétique , Transporteurs ABC/métabolisme , Plaque amyloïde/anatomopathologie , Plaque amyloïde/génétique , Plaque amyloïde/métabolisme , Peptides bêta-amyloïdes/métabolisme , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Neurones/métabolisme , Neurones/anatomopathologie , Modèles animaux de maladie humaine , Humains , Encéphale/anatomopathologie , Encéphale/métabolisme , Microglie/métabolisme , Microglie/anatomopathologie , Phagocytose/génétique , Précurseur de la protéine bêta-amyloïde/génétique
13.
Cell Rep Med ; 5(3): 101450, 2024 Mar 19.
Article de Anglais | MEDLINE | ID: mdl-38508139

RÉSUMÉ

CD47 is a ligand of SIRPα, an inhibitory receptor expressed by macrophages, dendritic cells, and natural killer (NK) cells, and, therefore, transgenic overexpression of CD47 is considered an effective approach to inhibiting transplant rejection. However, the detrimental effect of CD47 signaling is overlooked when exploring this approach. Here, we construct a mutant CD47 by replacing the transmembrane and intracellular domains with a membrane anchor (CD47-IgV). In both human and mouse cells, CD47-IgV is efficiently expressed on the cell surface and protects against phagocytosis in vitro and in vivo but does not induce cell death or inhibit angiogenesis. Furthermore, hematopoietic stem cells expressing transgenic CD47-IgV show no detectable alterations in engraftment or differentiation. This study provides a potentially effective means of achieving transgenic CD47 expression that may help to produce gene-edited pigs for xenotransplantation and hypoimmunogenic pluripotent stem cells for regenerative medicine.


Sujet(s)
, Antigènes CD47 , Animaux , Humains , Souris , Antigènes CD47/génétique , Antigènes CD47/métabolisme , Mort cellulaire , Phagocytose/génétique , Récepteurs immunologiques/génétique , Récepteurs immunologiques/métabolisme , Suidae
14.
J Neuroinflammation ; 21(1): 74, 2024 Mar 25.
Article de Anglais | MEDLINE | ID: mdl-38528525

RÉSUMÉ

The retinal pigment epithelium (RPE) maintains photoreceptor viability and function, completes the visual cycle, and forms the outer blood-retinal barrier (oBRB). Loss of RPE function gives rise to several monogenic retinal dystrophies and contributes to age-related macular degeneration. Retinal detachment (RD) causes separation of the neurosensory retina from the underlying RPE, disrupting the functional and metabolic relationships between these layers. Although the retinal response to RD is highly studied, little is known about how the RPE responds to loss of this interaction. RNA sequencing (RNA-Seq) was used to compare normal and detached RPE in the C57BL6/J mouse. The naïve mouse RPE transcriptome was compared to previously published RPE signature gene lists and from the union of these 14 genes (Bmp4, Crim1, Degs1, Gja1, Itgav, Mfap3l, Pdpn, Ptgds, Rbp1, Rnf13, Rpe65, Slc4a2, Sulf1 and Ttr) representing a core signature gene set applicable across rodent and human RPE was derived. Gene ontology enrichment analysis (GOEA) of the mouse RPE transcriptome identified expected RPE features and functions, such as pigmentation, phagocytosis, lysosomal and proteasomal degradation of proteins, and barrier function. Differentially expressed genes (DEG) at 1 and 7 days post retinal detachment (dprd) were defined as mRNA with a significant (padj≤0.05) fold change (FC) of 0.67 ≥ FC ≥ 1.5 in detached versus naïve RPE. The RPE transcriptome exhibited dramatic changes at 1 dprd, with 2297 DEG identified. The KEGG pathways and biological process GO groups related to innate immune responses were significantly enriched. Lipocalin 2 (Lcn2) and several chemokines were upregulated, while numerous genes related to RPE functions, such as pigment synthesis, visual cycle, phagocytosis, and tight junctions were downregulated at 1 dprd. The response was largely transient, with only 18 significant DEG identified at 7 dprd, including upregulation of complement gene C4b. Validation studies confirmed RNA-Seq results. Thus, the RPE quickly downregulates cell-specific functions and mounts an innate immune defense response following RD. Our data demonstrate that the RPE contributes to the inflammatory response to RD and may play a role in attraction of immune cells to the subretinal space.


Sujet(s)
Dégénérescence maculaire , Décollement de la rétine , Souris , Animaux , Humains , Épithélium pigmentaire de la rétine/métabolisme , Décollement de la rétine/métabolisme , Rétine/métabolisme , Dégénérescence maculaire/métabolisme , Phagocytose/génétique , Récepteurs de la protéine morphogénique osseuse/métabolisme
15.
Int Immunopharmacol ; 130: 111708, 2024 Mar 30.
Article de Anglais | MEDLINE | ID: mdl-38394889

RÉSUMÉ

Immunoevasion has been a severe obstacle for the clinical treatment of breast cancer (BC). CD47, known as an anti-phagocytic molecule, plays a key role in governing the evasion of tumor cells from immune surveillance by interacting with signal-regulated protein α (SIRPα) on macrophages. Here, we report for the first time that miR-299-3p is a direct regulator of CD47 with tumor suppressive effects both in vitro and in vivo. miRNA expression profiles and overall survival of BC cohorts from the Cancer Genome Atlas, METABRIC, or GSE19783 datasets showed that miR-299-3p is downregulated in BC tissues and that BC patients with low levels of miR-299-3p have poorer prognoses. Using dual-luciferase reporter, qRT-PCR, Western blot, and phagocytosis assays, we proved that restoration of miR-299-3p can suppress CD47 expression by directly targeting the predicted seed sequence "CCCACAU" in its 3'-UTR, leading to phagocytosis of BC cells by macrophages, whereas miR-299-3p inhibition or deletion reversed this effect. Additionally, Gene Ontology (GO) analysis and a variety of confirmatory experiments revealed that miR-299-3p was inversely correlated with cell proliferation, migration, and the cell cycle process. Mechanistically, miR-299-3p can also directly target ABCE1, an essential ribosome recycling factor, alleviating these malignant phenotypes of BC cells. In vivo BC xenografts based on nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice further proved that restoration of miR-299-3p resulted in a significant suppression of tumorigenesis and a promotion of macrophage activation and infiltration. Overall, our study suggested that miR-299-3p is a potent inhibitor of CD47 and ABCE1 to exhibit bifunctional BC-suppressing effects through immune activation conjugated with malignant behavior inhibition in breast carcinogenesis and thus can potentially serve as a novel therapeutic target for BC.


Sujet(s)
Transporteurs ABC , Tumeurs du sein , Antigènes CD47 , microARN , Échappement de la tumeur à la surveillance immunitaire , Animaux , Femelle , Humains , Souris , Transporteurs ABC/métabolisme , Tumeurs du sein/immunologie , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Carcinogenèse , Antigènes CD47/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Macrophages/métabolisme , Souris de lignée NOD , Souris SCID , microARN/métabolisme , Phagocytose/génétique , Phénotype
16.
Cell Rep ; 43(2): 113683, 2024 Feb 27.
Article de Anglais | MEDLINE | ID: mdl-38261512

RÉSUMÉ

Microglia are implicated as primarily detrimental in pain models; however, they exist across a continuum of states that contribute to homeostasis or pathology depending on timing and context. To clarify the specific contribution of microglia to pain progression, we take advantage of a temporally controlled transgenic approach to transiently deplete microglia. Unexpectedly, we observe complete resolution of pain coinciding with microglial repopulation rather than depletion. We find that repopulated mouse spinal cord microglia are morphologically distinct from control microglia and exhibit a unique transcriptome. Repopulated microglia from males and females express overlapping networks of genes related to phagocytosis and response to stress. We intersect the identified mouse genes with a single-nuclei microglial dataset from human spinal cord to identify human-relevant genes that may ultimately promote pain resolution after injury. This work presents a comprehensive approach to gene discovery in pain and provides datasets for the development of future microglial-targeted therapeutics.


Sujet(s)
Microglie , Transcriptome , Mâle , Femelle , Souris , Humains , Animaux , Transcriptome/génétique , Douleur/génétique , Douleur/anatomopathologie , Moelle spinale/anatomopathologie , Phagocytose/génétique
17.
Nat Rev Genet ; 25(6): 416-430, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38263430

RÉSUMÉ

Horizontal gene transfer (HGT), or lateral gene transfer, is the non-sexual movement of genetic information between genomes. It has played a pronounced part in bacterial and archaeal evolution, but its role in eukaryotes is less clear. Behaviours unique to eukaryotic cells - phagocytosis and endosymbiosis - have been proposed to increase the frequency of HGT, but nuclear genomes encode fewer HGTs than bacteria and archaea. Here, I review the existing theory in the context of the growing body of data on HGT in eukaryotes, which suggests that any increased chance of acquiring new genes through phagocytosis and endosymbiosis is offset by a reduced need for these genes in eukaryotes, because selection in most eukaryotes operates on variation not readily generated by HGT.


Sujet(s)
Transfert horizontal de gène , Eucaryotes/génétique , Symbiose/génétique , Cellules eucaryotes/métabolisme , Animaux , Phagocytose/génétique , Archéobactéries/génétique , Évolution moléculaire , Modèles génétiques
18.
Biochim Biophys Acta Mol Basis Dis ; 1870(2): 166978, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38061598

RÉSUMÉ

Phospholipase C-gamma 2 (PLCγ2) is highly expressed in hematopoietic and immune cells, where it is a key signalling node enabling diverse cellular functions. Within the periphery, gain-of-function (GOF) PLCγ2 variants, such as the strongly hypermorphic S707Y, cause severe immune dysregulation. The milder hypermorphic mutation PLCγ2 P522R increases longevity and confers protection in central nervous system (CNS) neurodegenerative disorders, implicating PLCγ2 as a novel therapeutic target for treating these CNS indications. Currently, nothing is known about what consequences strong PLCγ2 GOF has on CNS functionality, and more precisely on the specific biological functions of microglia. Using the PLCγ2 S707Y variant as a model of chronic activation we investigated the functional consequences of strong PLCγ2 GOF on human microglia. PLCγ2 S707Y expressing human inducible pluripotent stem cells (hiPSC)-derived microglia exhibited hypermorphic enzymatic activity under both basal and stimulated conditions, compared to PLCγ2 wild type. Despite the increase in PLCγ2 enzymatic activity, the PLCγ2 S707Y hiPSC-derived microglia display diminished functionality for key microglial processes including phagocytosis and cytokine secretion upon inflammatory challenge. RNA sequencing revealed a downregulation of genes related to innate immunity and response, providing molecular support for the phenotype observed. Our data suggests that chronic activation of PLCγ2 elicits a detrimental phenotype that is contributing to unfavourable CNS functions, and informs on the therapeutic window for targeting PLCγ2 in the CNS. Drug candidates targeting PLCγ2 will need to precisely mimic the effects of the PLCγ2 P522R variant on microglial function, but not those of the PLCγ2 S707Y variant.


Sujet(s)
Microglie , Maladies neurodégénératives , Humains , Encéphale/métabolisme , Immunité innée , Microglie/métabolisme , Maladies neurodégénératives/métabolisme , Phagocytose/génétique , Phospholipase C gamma/génétique , Phospholipase C gamma/métabolisme , Phospholipase C gamma/pharmacologie
19.
Exp Biol Med (Maywood) ; 248(23): 2289-2303, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-38062999

RÉSUMÉ

Genome-wide CRISPR-Cas9 knockout screens have emerged as a powerful method for identifying key genes driving tumor growth. The aim of this study was to explore the phagocytosis regulators (PRs) specifically associated with lower-grade glioma (LGG) using the CRISPR-Cas9 screening database. Identifying these core PRs could lead to novel therapeutic targets and pave the way for a non-invasive radiogenomics approach to assess LGG patients' prognosis and treatment response. We selected 24 PRs that were overexpressed and lethal in LGG for analysis. The identified PR subtypes (PRsClusters, geneClusters, and PRs-score models) effectively predicted clinical outcomes in LGG patients. Immune response markers, such as CTLA4, were found to be significantly associated with PR-score. Nine radiogenomics models using various machine learning classifiers were constructed to uncover survival risk. The area under the curve (AUC) values for these models in the test and training datasets were 0.686 and 0.868, respectively. The CRISPR-Cas9 screen identified novel prognostic radiogenomics biomarkers that correlated well with the expression status of specific PR-related genes in LGG patients. These biomarkers successfully stratified patient survival outcomes and treatment response using The Cancer Genome Atlas (TCGA) database. This study has important implications for the development of precise clinical treatment strategies and holds promise for more accurate therapeutic approaches for LGG patients in the future.


Sujet(s)
Tumeurs du cerveau , Gliome , Humains , Gliome/génétique , Aire sous la courbe , Bases de données factuelles , Apprentissage machine , Phagocytose/génétique , Tumeurs du cerveau/génétique
20.
Parasites Hosts Dis ; 61(4): 397-404, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-38043535

RÉSUMÉ

Acanthamoeba species are free-living amoebae those are widely distributed in the environment. They feed on various microorganisms, including bacteria, fungi, and algae. Although majority of the microbes phagocytosed by Acanthamoeba spp. are digested, some pathogenic bacteria thrive within them. Here, we identified the roles of 3 phagocytosis-associated genes (ACA1_077100, ACA1_175060, and AFD36229.1) in A. castellanii. These 3 genes were upregulated after the ingestion of Escherichia coli. However, after the ingestion of Legionella pneumophila, the expression of these 3 genes was not altered after the consumption of L. pneumophila. Furthermore, A. castellanii transfected with small interfering RNS (siRNA) targeting the 3 phagocytosis-associated genes failed to digest phagocytized E. coli. Silencing of ACA1_077100 disabled phagosome formation in the E. coli-ingesting A. castellanii. Alternatively, silencing of ACA1_175060 enabled phagosome formation; however, phagolysosome formation was inhibited. Moreover, suppression of AFD36229.1 expression prevented E. coli digestion and consequently led to the rupturing of A. castellanii. Our results demonstrated that the ACA1_077100, ACA1_175060, and AFD36229.1 genes of Acanthamoeba played crucial roles not only in the formation of phagosome and phagolysosome but also in the digestion of E. coli.


Sujet(s)
Acanthamoeba castellanii , Legionella pneumophila , Acanthamoeba castellanii/génétique , Escherichia coli/génétique , Phagocytose/génétique , Phagosomes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE