Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.226
Filtrer
1.
Front Immunol ; 15: 1386658, 2024.
Article de Anglais | MEDLINE | ID: mdl-39104537

RÉSUMÉ

Alcohol ingestion is a widespread habituation that evolved along with a growing population, altering physiological conditions through immunomodulatory function. There is much research that has reported that consumption of alcohol at low and heavy levels causes different biological impacts, including cellular injury, leading to systemic dysfunction and increased inflammatory markers. In the fate of professional phagocytic cells, efferocytosis is an inevitable mechanism activated by the apoptotic cells, thus eliminating them and preventing the accumulation of cell corpses/debris in the microenvironment. Subsequently, it promotes the tissue repair mechanism and maintains cellular homeostasis. Unfortunately, defective efferocytosis is widely found in several inflammatory and age-related diseases such as atherosclerosis, autoimmune diseases, lung injury, fatty liver disease, and neurodegenerative diseases. Alcohol abuse is one of the factors that provoke an immune response that increases the rate of morbidity and mortality in parallel in systemic disease patients. Information regarding the emergence of immunomodulation during alcoholic pathogenesis and its association with efferocytosis impairment remain elusive. Hence, here in this review, we discussed the mechanism of efferocytosis, the role of defective efferocytosis in inflammatory diseases, and the role of alcohol on efferocytosis impairment.


Sujet(s)
Intoxication alcoolique , , Animaux , Humains , Intoxication alcoolique/immunologie , Intoxication alcoolique/métabolisme , Apoptose , /immunologie , Éthanol , Inflammation/immunologie , Macrophages/immunologie , Macrophages/métabolisme , Phagocytes/immunologie , Phagocytes/métabolisme
2.
Front Immunol ; 15: 1389674, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994369

RÉSUMÉ

Cell death is an important process in the body, as it occurs throughout every tissue during development, disease, and tissue regeneration. Phagocytes are responsible for clearing away dying cells and are typically characterized as either professional or nonprofessional phagocytes. Professional phagocytes, such as macrophages, are found in nearly every part of the body while nonprofessional phagocytes, such as epithelial cells, are found in every tissue type. However, there are organs that are considered "immune-privileged" as they have little to no immune surveillance and rely on nonprofessional phagocytes to engulf dying cells. These organs are surrounded by barriers to protect the tissue from viruses, bacteria, and perhaps even immune cells. The Drosophila ovary is considered immune-privileged, however the presence of hemocytes, the macrophages of Drosophila, around the ovary suggests they may have a potential function. Here we analyze hemocyte localization and potential functions in response to starvation-induced cell death in the ovary. Hemocytes were found to accumulate in the oviduct in the vicinity of mature eggs and follicle cell debris. Genetic ablation of hemocytes revealed that the presence of hemocytes affects oogenesis and that they phagocytose ovarian cell debris and in their absence fecundity decreases. Unpaired3, an IL-6 like cytokine, was found to be required for the recruitment of hemocytes to the oviduct to clear away obsolete follicle cells. These findings demonstrate a role for hemocytes in the ovary, providing a more thorough understanding of phagocyte communication and cell clearance in a previously thought immune-privileged organ.


Sujet(s)
Hémocytes , Ovaire , Phagocytes , Phagocytose , Animaux , Femelle , Ovaire/immunologie , Hémocytes/immunologie , Phagocytes/immunologie , Phagocytes/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Drosophila melanogaster/immunologie , Ovogenèse , Drosophila/immunologie
3.
Sci Rep ; 14(1): 14187, 2024 06 20.
Article de Anglais | MEDLINE | ID: mdl-38902328

RÉSUMÉ

Mononuclear phagocytes (MNP), including macrophages and dendritic cells form an essential component of primary responses to environmental hazards and toxic exposures. This is particularly important in disease conditions such as asthma and allergic airway disease, where many different cell types are present. In this study, we differentiated CD34+ haematopoietic stem cells towards different populations of MNP in an effort to understand how different cell subtypes present in inflammatory disease microenvironments respond to the common allergen house dust mite (HDM). Using single cell mRNA sequencing, we demonstrate that macrophage subtypes MCSPP1+ and MLCMARCO+ display different patterns of gene expression after HDM challenge, noted especially for the chemokines CXCL5, CXCL8, CCL5 and CCL15. MLCCD206Hi alternatively activated macrophages displayed the greatest changes in expression, while neutrophil and monocyte populations did not respond. Further work investigated how pollutant diesel exhaust particles could modify these transcriptional responses and revealed that CXC but not CC type chemokines were further upregulated. Through the use of diesel particles with adsorbed material removed, we suggest that soluble pollutants on these particles are the active constituents responsible for the modifying effects on HDM. This study highlights that environmental exposures may influence tissue responses dependent on which MNP cell type is present, and that these should be considerations when modelling such events in vitro. Understanding the nuanced responsiveness of different immune cell types to allergen and pollutant exposure also contributes to a better understanding of how these exposures influence the development and exacerbation of human disease.


Sujet(s)
Pyroglyphidae , Animaux , Pyroglyphidae/immunologie , Humains , Phagocytes/métabolisme , Phagocytes/immunologie , Macrophages/métabolisme , Macrophages/immunologie , Allergènes/immunologie , Emissions des véhicules/toxicité , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/immunologie , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques
4.
Proc Natl Acad Sci U S A ; 121(25): e2312499121, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38857395

RÉSUMÉ

Ex vivo expansion of human CD34+ hematopoietic stem and progenitor cells remains a challenge due to rapid differentiation after detachment from the bone marrow niche. In this study, we assessed the capacity of an inducible fusion protein to enable sustained ex vivo proliferation of hematopoietic precursors and their capacity to differentiate into functional phagocytes. We fused the coding sequences of an FK506-Binding Protein 12 (FKBP12)-derived destabilization domain (DD) to the myeloid/lymphoid lineage leukemia/eleven nineteen leukemia (MLL-ENL) fusion gene to generate the fusion protein DD-MLL-ENL and retrovirally expressed the protein switch in human CD34+ progenitors. Using Shield1, a chemical inhibitor of DD fusion protein degradation, we established large-scale and long-term expansion of late monocytic precursors. Upon Shield1 removal, the cells lost self-renewal capacity and spontaneously differentiated, even after 2.5 y of continuous ex vivo expansion. In the absence of Shield1, stimulation with IFN-γ, LPS, and GM-CSF triggered terminal differentiation. Gene expression analysis of the obtained phagocytes revealed marked similarity with naïve monocytes. In functional assays, the novel phagocytes migrated toward CCL2, attached to VCAM-1 under shear stress, produced reactive oxygen species, and engulfed bacterial particles, cellular particles, and apoptotic cells. Finally, we demonstrated Fcγ receptor recognition and phagocytosis of opsonized lymphoma cells in an antibody-dependent manner. Overall, we have established an engineered protein that, as a single factor, is useful for large-scale ex vivo production of human phagocytes. Such adjustable proteins have the potential to be applied as molecular tools to produce functional immune cells for experimental cell-based approaches.


Sujet(s)
Différenciation cellulaire , Phagocytes , Humains , Phagocytes/métabolisme , Cellules souches hématopoïétiques/métabolisme , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/génétique , Protéine de la leucémie myéloïde-lymphoïde/métabolisme , Protéine de la leucémie myéloïde-lymphoïde/génétique , Leucémies/génétique , Leucémies/anatomopathologie , Leucémies/métabolisme , Ingénierie des protéines/méthodes , Phagocytose
5.
Sci Adv ; 10(23): eadl6083, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38838151

RÉSUMÉ

Hepatocellular carcinoma (HCC) acquires an immunosuppressive microenvironment, leading to unbeneficial therapeutic outcomes. Hyaluronan-mediated motility receptor (HMMR) plays a crucial role in tumor progression. Here, we found that aberrant expression of HMMR could be a predictive biomarker for the immune suppressive microenvironment of HCC, but the mechanism remains unclear. We established an HMMR-/- liver cancer mouse model to elucidate the HMMR-mediated mechanism of the dysregulated "don't eat me" signal. HMMR knockout inhibited liver cancer growth and induced phagocytosis. HMMRhigh liver cancer cells escaped from phagocytosis via sustaining CD47 signaling. Patients with HMMRhighCD47high expression showed a worse prognosis than those with HMMRlowCD47low expression. HMMR formed a complex with FAK/SRC in the cytoplasm to activate NF-κB signaling, which could be independent of membrane interaction with CD44. Notably, targeting HMMR could enhance anti-PD-1 treatment efficiency by recruiting CD8+ T cells. Overall, our data revealed a regulatory mechanism of the "don't eat me" signal and knockdown of HMMR for enhancing anti-PD-1 treatment.


Sujet(s)
Antigènes CD47 , Carcinome hépatocellulaire , Antigènes CD44 , Tumeurs du foie , Phagocytes , Phagocytose , Animaux , Humains , Souris , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/génétique , Antigènes CD47/métabolisme , Antigènes CD47/génétique , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Focal adhesion kinase 1/métabolisme , Focal adhesion kinase 1/génétique , Antigènes CD44/métabolisme , Antigènes CD44/génétique , Échappement immunitaire , Tumeurs du foie/anatomopathologie , Tumeurs du foie/immunologie , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Souris knockout , Facteur de transcription NF-kappa B/métabolisme , Phagocytes/métabolisme , Phagocytes/immunologie , Transduction du signal , Échappement de la tumeur à la surveillance immunitaire , Microenvironnement tumoral/immunologie
6.
Carbohydr Polym ; 339: 122256, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38823922

RÉSUMÉ

Recently, the intestinal lymphatic transport based on Peyer's patches (PPs) is emerging as a promising absorption pathway for natural polysaccharides. Herein, the aim of this study is to investigate the PP-based oral absorption of a pectic polysaccharide from Smilax china L. (SCLP), as well as its uptake and transport mechanisms in related immune cells. Taking advantages of the traceability of fluorescently labeled SCLP, we confirmed that SCLP could be absorbed into PPs and captured by their mononuclear phagocytes (dendritic cells and macrophages) following oral administration. Subsequently, the systematic in vitro study suggested that the endocytic mechanisms of SCLP by model mononuclear phagocytes (BMDCs and RAW264.7 cells) mainly involved caveolae-mediated endocytosis, macropinocytosis and phagocytosis. More importantly, SCLP directly binds and interacts with toll-like receptor 2 (TLR2) and galectin 3 (Gal-3) receptor, and was taken up by mononuclear phagocytes in receptor-mediated manner. After internalization, SCLP was intracellularly transported primarily through endolysosomal pathway and ultimately localized in lysosomes. In summary, this work reveals novel information and perspectives about the in vivo fate of SCLP, which will contribute to further research and utilization of SCLP and other pectic polysaccharides.


Sujet(s)
Plaques de Peyer , Smilax , Animaux , Souris , Cellules RAW 264.7 , Plaques de Peyer/métabolisme , Smilax/composition chimique , Endocytose , Pectine/composition chimique , Pectine/métabolisme , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Phagocytose/effets des médicaments et des substances chimiques , Phagocytes/métabolisme , Phagocytes/effets des médicaments et des substances chimiques , Récepteur de type Toll-2/métabolisme , Souris de lignée BALB C , Mâle , Cellules dendritiques/métabolisme , Cellules dendritiques/effets des médicaments et des substances chimiques , Administration par voie orale
7.
Methods Mol Biol ; 2813: 39-64, 2024.
Article de Anglais | MEDLINE | ID: mdl-38888769

RÉSUMÉ

One hundred years have passed since the death of Élie Metchnikoff (1845-1916). He was the first to observe the uptake of particles by cells and realized the importance of this process, named phagocytosis, for the host response to injury and infection. He also was a strong advocate of the role of phagocytosis in cellular immunity, and with this, he gave us the basis for our modern understanding of inflammation and the innate immune response. Phagocytosis is an elegant but complex process for the ingestion and elimination of pathogens, but it is also important for the elimination of apoptotic cells and hence fundamental for tissue homeostasis. Phagocytosis can be divided into four main steps: (i) recognition of the target particle, (ii) signaling to activate the internalization machinery, (iii) phagosome formation, and (iv) phagolysosome maturation. In this chapter, we present a general view of our current knowledge on phagocytosis performed mainly by professional phagocytes through antibody and complement receptors and discuss aspects that remain incompletely understood.


Sujet(s)
Phagocytose , Phagosomes , Humains , Animaux , Phagosomes/métabolisme , Phagocytes/immunologie , Phagocytes/métabolisme , Transduction du signal , Immunité innée
8.
mBio ; 15(6): e0086224, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38767353

RÉSUMÉ

Mammalian target of rapamycin (mTOR) is a key regulator of metabolism in the mammalian cell. Here, we show the essential role for mTOR signaling in the immune response to bacterial infection. Inhibition of mTOR during infection with Staphylococcus aureus revealed that mTOR signaling is required for bactericidal free radical production by phagocytes. Mechanistically, mTOR supported glucose transporter GLUT1 expression, potentially through hypoxia-inducible factor 1α, upon phagocyte activation. Cytokine and chemokine signaling, inducible nitric oxide synthase, and p65 nuclear translocation were present at similar levels during mTOR suppression, suggesting an NF-κB-independent role for mTOR signaling in the immune response during bacterial infection. We propose that mTOR signaling primarily mediates the metabolic requirements necessary for phagocyte bactericidal free radical production. This study has important implications for the metabolic requirements of innate immune cells during bacterial infection as well as the clinical use of mTOR inhibitors.IMPORTANCESirolimus, everolimus, temsirolimus, and similar are a class of pharmaceutics commonly used in the clinical treatment of cancer and the anti-rejection of transplanted organs. Each of these agents suppresses the activity of the mammalian target of rapamycin (mTOR), a master regulator of metabolism in human cells. Activation of mTOR is also involved in the immune response to bacterial infection, and treatments that inhibit mTOR are associated with increased susceptibility to bacterial infections in the skin and soft tissue. Infections caused by Staphylococcus aureus are among the most common and severe. Our study shows that this susceptibility to S. aureus infection during mTOR suppression is due to an impaired function of phagocytic immune cells responsible for controlling bacterial infections. Specifically, we observed that mTOR activity is required for phagocytes to produce antimicrobial free radicals. These results have important implications for immune responses during clinical treatments and in disease states where mTOR is suppressed.


Sujet(s)
Transporteur de glucose de type 1 , Phagocytes , Transduction du signal , Infections à staphylocoques , Staphylococcus aureus , Sérine-thréonine kinases TOR , Staphylococcus aureus/immunologie , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Infections à staphylocoques/immunologie , Infections à staphylocoques/microbiologie , Phagocytes/immunologie , Phagocytes/métabolisme , Phagocytes/microbiologie , Humains , Transporteur de glucose de type 1/métabolisme , Transporteur de glucose de type 1/génétique , Animaux , Radicaux libres/métabolisme , Souris , Souris de lignée C57BL
9.
Front Immunol ; 15: 1372904, 2024.
Article de Anglais | MEDLINE | ID: mdl-38742116

RÉSUMÉ

Introduction: The California purple sea urchin, Strongylocentrotus purpuratus, relies solely on an innate immune system to combat the many pathogens in the marine environment. One aspect of their molecular defenses is the SpTransformer (SpTrf) gene family that is upregulated in response to immune challenge. The gene sequences are highly variable both within and among animals and likely encode thousands of SpTrf isoforms within the sea urchin population. The native SpTrf proteins bind foreign targets and augment phagocytosis of a marine Vibrio. A recombinant (r)SpTrf-E1-Ec protein produced by E. coli also binds Vibrio but does not augment phagocytosis. Methods: To address the question of whether other rSpTrf isoforms function as opsonins and augment phagocytosis, six rSpTrf proteins were expressed in insect cells. Results: The rSpTrf proteins are larger than expected, are glycosylated, and one dimerized irreversibly. Each rSpTrf protein cross-linked to inert magnetic beads (rSpTrf::beads) results in different levels of surface binding and phagocytosis by phagocytes. Initial analysis shows that significantly more rSpTrf::beads associate with cells compared to control BSA::beads. Binding specificity was verified by pre-incubating the rSpTrf::beads with antibodies, which reduces the association with phagocytes. The different rSpTrf::beads show significant differences for cell surface binding and phagocytosis by phagocytes. Furthermore, there are differences among the three distinct types of phagocytes that show specific vs. constitutive binding and phagocytosis. Conclusion: These findings illustrate the complexity and effectiveness of the sea urchin innate immune system driven by the natSpTrf proteins and the phagocyte cell populations that act to neutralize a wide range of foreign pathogens.


Sujet(s)
Phagocytes , Phagocytose , Protéines recombinantes , Animaux , Phagocytose/immunologie , Phagocytes/immunologie , Phagocytes/métabolisme , Protéines recombinantes/immunologie , Protéines recombinantes/métabolisme , Protéines recombinantes/génétique , Liaison aux protéines , Strongylocentrotus purpuratus/immunologie , Strongylocentrotus purpuratus/génétique , Immunité innée , Isoformes de protéines/génétique , Isoformes de protéines/immunologie , Echinoidea/immunologie , Vibrio/immunologie , Opsonines/métabolisme , Opsonines/immunologie
10.
Cells ; 13(10)2024 May 11.
Article de Anglais | MEDLINE | ID: mdl-38786045

RÉSUMÉ

Macrophages and microglia are professional phagocytes that sense and migrate toward "eat-me" signals. The role of phagocytic cells is to maintain homeostasis by engulfing senescent or apoptotic cells, debris, and abnormally aggregated macromolecules. Usually, dying cells send out "find-me" signals, facilitating the recruitment of phagocytes. Healthy cells can also promote or inhibit the phagocytosis phenomenon of macrophages and microglia by tuning the balance between "eat-me" and "don't-eat-me" signals at different stages in their lifespan, while the "don't-eat-me" signals are often hijacked by tumor cells as a mechanism of immune evasion. Using a combination of bioinformatic analysis and spatial profiling, we delineate the balance of the "don't-eat-me" CD47/SIRPα and "eat-me" CALR/STC1 ligand-receptor interactions to guide therapeutic strategies that are being developed for glioblastoma sequestered in the central nervous system (CNS).


Sujet(s)
Antigènes CD47 , Calréticuline , Glioblastome , Phagocytes , Phagocytose , Humains , Glioblastome/anatomopathologie , Glioblastome/thérapie , Glioblastome/métabolisme , Antigènes CD47/métabolisme , Phagocytes/métabolisme , Calréticuline/métabolisme , Récepteurs immunologiques/métabolisme , Macrophages/métabolisme , Macrophages/immunologie , Microglie/métabolisme , Microglie/anatomopathologie , Mort cellulaire , Animaux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/thérapie , Antigènes de différenciation
11.
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 ,
12.
EMBO Rep ; 25(6): 2550-2570, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38730209

RÉSUMÉ

Transmembrane protein 268 (TMEM268) is a novel, tumor growth-related protein first reported by our laboratory. It interacts with the integrin subunit ß4 (ITGB4) and plays a positive role in the regulation of the ITGB4/PLEC signaling pathway. Here, we investigated the effects and mechanism of TMEM268 in anti-infectious immune response in mice. Tmem268 knockout in mice aggravated cecal ligation and puncture-induced sepsis, as evidenced by higher bacterial burden in various tissues and organs, congestion, and apoptosis. Moreover, Tmem268 deficiency in mice inhibited phagocyte adhesion and migration, thus decreasing phagocyte infiltration at the site of infection and complement-dependent phagocytosis. Further findings indicated that TMEM268 interacts with CD11b and inhibits its degradation via the endosome-lysosome pathway. Our results reveal a positive regulatory role of TMEM268 in ß2 integrin-associated anti-infectious immune responses and signify the potential value of targeting the TMEM268-CD11b signaling axis for the maintenance of immune homeostasis and immunotherapy for sepsis and related immune disorders.


Sujet(s)
Antigènes CD11b , Protéines membranaires , Souris knockout , Sepsie , Transduction du signal , Animaux , Humains , Souris , Antigènes CD11b/métabolisme , Antigènes CD11b/génétique , Adhérence cellulaire/génétique , Mouvement cellulaire/génétique , Régulation négative , Endosomes/métabolisme , Délétion de gène , Lysosomes/métabolisme , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Souris de lignée C57BL , Phagocytes/métabolisme , Phagocytes/immunologie , Phagocytose , Sepsie/génétique , Sepsie/immunologie , Sepsie/métabolisme
13.
Proc Natl Acad Sci U S A ; 121(23): e2320388121, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38805284

RÉSUMÉ

Essential for reactive oxygen species (EROS) protein is a recently identified molecular chaperone of NOX2 (gp91phox), the catalytic subunit of phagocyte NADPH oxidase. Deficiency in EROS is a recently identified cause for chronic granulomatous disease, a genetic disorder with recurrent bacterial and fungal infections. Here, we report a cryo-EM structure of the EROS-NOX2-p22phox heterotrimeric complex at an overall resolution of 3.56Å. EROS and p22phox are situated on the opposite sides of NOX2, and there is no direct contact between them. EROS associates with NOX2 through two antiparallel transmembrane (TM) α-helices and multiple ß-strands that form hydrogen bonds with the cytoplasmic domain of NOX2. EROS binding induces a 79° upward bend of TM2 and a 48° backward rotation of the lower part of TM6 in NOX2, resulting in an increase in the distance between the two hemes and a shift of the binding site for flavin adenine dinucleotide (FAD). These conformational changes are expected to compromise superoxide production by NOX2, suggesting that the EROS-bound NOX2 is in a protected state against activation. Phorbol myristate acetate, an activator of NOX2 in vitro, is able to induce dissociation of NOX2 from EROS with concurrent increase in FAD binding and superoxide production in a transfected COS-7 model. In differentiated neutrophil-like HL-60, the majority of NOX2 on the cell surface is dissociated with EROS. Further studies are required to delineate how EROS dissociates from NOX2 during its transport to cell surface, which may be a potential mechanism for regulation of NOX2 activation.


Sujet(s)
Cryomicroscopie électronique , NADPH Oxidase 2 , NADPH oxidase , Phagocytes , Humains , NADPH Oxidase 2/métabolisme , NADPH Oxidase 2/génétique , NADPH Oxidase 2/composition chimique , Phagocytes/métabolisme , NADPH oxidase/métabolisme , NADPH oxidase/génétique , NADPH oxidase/composition chimique , Liaison aux protéines , Sites de fixation , Granulomatose septique chronique/métabolisme , Granulomatose septique chronique/génétique , Modèles moléculaires , Espèces réactives de l'oxygène/métabolisme
14.
Am J Physiol Endocrinol Metab ; 327(1): E55-E68, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38717364

RÉSUMÉ

Statins are used to treat hypercholesterolemia and function by inhibiting the production of the rate-limiting metabolite mevalonate. As such, statin treatment not only inhibits de novo synthesis of cholesterol but also isoprenoids that are involved in prenylation, the posttranslational lipid modification of proteins. The immunomodulatory effects of statins are broad and often conflicting. Previous work demonstrated that statins increased survival and inhibited myeloid cell trafficking in a murine model of sepsis, but the exact mechanisms underlying this phenomenon were unclear. Herein, we investigated the role of prenylation in chemoattractant responses. We found that simvastatin treatment abolished chemoattractant responses induced by stimulation by C5a and FMLP. The inhibitory effect of simvastatin treatment was unaffected by the addition of either farnesyl pyrophosphate (FPP) or squalene but was reversed by restoring geranylgeranyl pyrophosphate (GGPP). Treatment with prenyltransferase inhibitors showed that the chemoattractant response to both chemoattractants was dependent on geranylgeranylation. Proteomic analysis of C15AlkOPP-prenylated proteins identified several geranylgeranylated proteins involved in chemoattractant responses, including RHOA, RAC1, CDC42, and GNG2. Chemoattractant responses in THP-1 human macrophages were also geranylgeranylation dependent. These studies provide data that help clarify paradoxical findings on the immunomodulatory effects of statins. Furthermore, they establish the role of geranylgeranylation in mediating the morphological response to chemoattractant C5a.NEW & NOTEWORTHY The immunomodulatory effect of prenylation is ill-defined. We investigated the role of prenylation on the chemoattractant response to C5a. Simvastatin treatment inhibits the cytoskeletal remodeling associated with a chemotactic response. We showed that the chemoattractant response to C5a was dependent on geranylgeranylation, and proteomic analysis identified several geranylgeranylated proteins that are involved in C5a receptor signaling and cytoskeletal remodeling. Furthermore, they establish the role of geranylgeranylation in mediating the response to chemoattractant C5a.


Sujet(s)
Polyisoprényl-phosphates , Polyisoprényl-phosphates/pharmacologie , Polyisoprényl-phosphates/métabolisme , Humains , Simvastatine/pharmacologie , Facteurs chimiotactiques/pharmacologie , Facteurs chimiotactiques/métabolisme , Phagocytes/effets des médicaments et des substances chimiques , Phagocytes/métabolisme , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Complément C5a/métabolisme , Prénylation des protéines/effets des médicaments et des substances chimiques , Animaux , Souris , Sesquiterpènes
15.
Methods Mol Biol ; 2775: 195-209, 2024.
Article de Anglais | MEDLINE | ID: mdl-38758319

RÉSUMÉ

Cryptococcus neoformans, the predominant etiological agent of cryptococcosis, is an encapsulated fungal pathogen found ubiquitously in the environment that causes pneumonia and life-threatening infections of the central nervous system. Following inhalation of yeasts or desiccated basidiospores into the lung alveoli, resident pulmonary phagocytic cells aid in the identification and eradication of Cryptococcus yeast through their arsenal of pattern recognition receptors (PRRs). PRRs recognize conserved pathogen-associated molecular patterns (PAMPs), such as branched mannans, ß-glucans, and chitins that are the major components of the fungal cell wall. However, the key receptors/ligand interactions required for cryptococcal recognition and eventual fungal clearance have yet to be elucidated. Here we present an imaging flow cytometer (IFC) method that offers a novel quantitative cellular imaging and population statistics tool to accurately measure phagocytosis of fungal cells. It has the capacity to measure two distinct steps of phagocytosis: association/attachment and internalization in a high-throughput and quantitative manner that is difficult to achieve with other technologies. Results from these IFC studies allow for the potential to identify PRRs required for recognition, uptake, and subsequent activation of cytokine production, as well as other effector cell responses required for fungal clearance.


Sujet(s)
Cryptococcus neoformans , Cytométrie en flux , Phagocytose , Cytométrie en flux/méthodes , Cryptococcus neoformans/métabolisme , Animaux , Souris , Phagocytes/métabolisme , Phagocytes/microbiologie , Cryptococcose/microbiologie , Cryptococcose/métabolisme , Cryptococcose/immunologie , Cryptococcus/métabolisme , Humains , Cytométrie en images/méthodes , Récepteurs de reconnaissance de motifs moléculaires/métabolisme
16.
Front Immunol ; 15: 1401294, 2024.
Article de Anglais | MEDLINE | ID: mdl-38720899

RÉSUMÉ

Inhibitory natural killer (NK) cell receptors recognize MHC class I (MHC-I) in trans on target cells and suppress cytotoxicity. Some NK cell receptors recognize MHC-I in cis, but the role of this interaction is uncertain. Ly49Q, an atypical Ly49 receptor expressed in non-NK cells, binds MHC-I in cis and mediates chemotaxis of neutrophils and type I interferon production by plasmacytoid dendritic cells. We identified a lipid-binding motif in the juxtamembrane region of Ly49Q and found that Ly49Q organized functional membrane domains comprising sphingolipids via sulfatide binding. Ly49Q recruited actin-remodeling molecules to an immunoreceptor tyrosine-based inhibitory motif, which enabled the sphingolipid-enriched membrane domain to mediate complicated actin remodeling at the lamellipodia and phagosome membranes during phagocytosis. Thus, Ly49Q facilitates integrative regulation of proteins and lipid species to construct a cell type-specific membrane platform. Other Ly49 members possess lipid binding motifs; therefore, membrane platform organization may be a primary role of some NK cell receptors.


Sujet(s)
Sphingolipides , Animaux , Humains , Sphingolipides/métabolisme , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Phagocytose , Phagocytes/immunologie , Phagocytes/métabolisme , Sous-famille A des récepteurs de cellules NK de type lectine/métabolisme , Membrane cellulaire/métabolisme , Liaison aux protéines
17.
Sci Signal ; 17(834): eadq0353, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38687824

RÉSUMÉ

Displacement of the glycocalyx by membrane blebbing enables macrophages to recognize apoptotic cells.


Sujet(s)
Apoptose , Glycocalyx , Macrophages , Humains , Glycocalyx/métabolisme , Animaux , Macrophages/métabolisme , Macrophages/cytologie , Phagocytes/métabolisme , Phagocytes/cytologie , Phagocytose , Souris
18.
Cell Rep Med ; 5(5): 101528, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38677283

RÉSUMÉ

Stimulator of interferon genes (STING)-dependent signaling is requisite for effective anti-microbial and anti-tumor activity. STING signaling is commonly defective in cancer cells, which enables tumor cells to evade the immunosurveillance system. We evaluate here whether intrinsic STING signaling in such tumor cells could be reconstituted by creating recombinant herpes simplex viruses (rHSVs) that express components of the STING signaling pathway. We observe that rHSVs expressing STING and/or cGAS replicate inefficiently yet retain in vivo anti-tumor activity, independent of oncolytic activity requisite on the trans-activation of extrinsic STING signaling in phagocytes by engulfed microbial dsDNA species. Accordingly, the in vivo effects of virotherapy could be simulated by nanoparticles incorporating non-coding dsDNA species, which comparably elicit the trans-activation of phagocytes and augment the efficacy of established cancer treatments including checkpoint inhibition and radiation therapy. Our results help elucidate mechanisms of virotherapeutic anti-tumor activity as well as provide alternate strategies to treat cancer.


Sujet(s)
ADN , Phagocytes , Animaux , Phagocytes/immunologie , Phagocytes/métabolisme , Humains , Souris , ADN/métabolisme , ADN/immunologie , ADN/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Transduction du signal , Nucleotidyltransferases/métabolisme , Nucleotidyltransferases/génétique , Lignée cellulaire tumorale , Tumeurs/immunologie , Tumeurs/anatomopathologie , Tumeurs/thérapie , Tumeurs/génétique , Simplexvirus/génétique , Simplexvirus/immunologie , Souris de lignée C57BL , Thérapie virale de cancers/méthodes
19.
mBio ; 15(5): e0342923, 2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38624208

RÉSUMÉ

The Hippo kinases MST1 and MST2 initiate a highly conserved signaling cascade called the Hippo pathway that limits organ size and tumor formation in animals. Intriguingly, pathogens hijack this host pathway during infection, but the role of MST1/2 in innate immune cells against pathogens is unclear. In this report, we generated Mst1/2 knockout macrophages to investigate the regulatory activities of the Hippo kinases in immunity. Transcriptomic analyses identified differentially expressed genes (DEGs) regulated by MST1/2 that are enriched in biological pathways, such as systemic lupus erythematosus, tuberculosis, and apoptosis. Surprisingly, pharmacological inhibition of the downstream components LATS1/2 in the canonical Hippo pathway did not affect the expression of a set of immune DEGs, suggesting that MST1/2 control these genes via alternative inflammatory Hippo signaling. Moreover, MST1/2 may affect immune communication by influencing the release of cytokines, including TNFα, CXCL10, and IL-1ra. Comparative analyses of the single- and double-knockout macrophages revealed that MST1 and MST2 differentially regulate TNFα release and expression of the immune transcription factor MAF, indicating that the two homologous Hippo kinases individually play a unique role in innate immunity. Notably, both MST1 and MST2 can promote apoptotic cell death in macrophages upon stimulation. Lastly, we demonstrate that the Hippo kinases are critical factors in mammalian macrophages and single-cell amoebae to restrict infection by Legionella pneumophila, Escherichia coli, and Pseudomonas aeruginosa. Together, these results uncover non-canonical inflammatory Hippo signaling in macrophages and the evolutionarily conserved role of the Hippo kinases in the anti-microbial defense of eukaryotic hosts. IMPORTANCE: Identifying host factors involved in susceptibility to infection is fundamental for understanding host-pathogen interactions. Clinically, individuals with mutations in the MST1 gene which encodes one of the Hippo kinases experience recurrent infection. However, the impact of the Hippo kinases on innate immunity remains largely undetermined. This study uses mammalian macrophages and free-living amoebae with single- and double-knockout in the Hippo kinase genes and reveals that the Hippo kinases are the evolutionarily conserved determinants of host defense against microbes. In macrophages, the Hippo kinases MST1 and MST2 control immune activities at multiple levels, including gene expression, immune cell communication, and programmed cell death. Importantly, these activities controlled by MST1 and MST2 in macrophages are independent of the canonical Hippo cascade that is known to limit tissue growth and tumor formation. Together, these findings unveil a unique inflammatory Hippo signaling pathway that plays an essential role in innate immunity.


Sujet(s)
Voie de signalisation Hippo , Immunité innée , Macrophages , Protein-Serine-Threonine Kinases , Serine-threonine kinase-3 , Transduction du signal , Animaux , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Souris , Macrophages/immunologie , Macrophages/microbiologie , Macrophages/métabolisme , Phagocytes/immunologie , Phagocytes/microbiologie , Phagocytes/métabolisme , Souris knockout , Infections bactériennes/immunologie , Infections bactériennes/microbiologie , Infections bactériennes/génétique , Analyse de profil d'expression de gènes , Souris de lignée C57BL , Pseudomonas aeruginosa/immunologie
20.
J Leukoc Biol ; 115(6): 1165-1176, 2024 05 29.
Article de Anglais | MEDLINE | ID: mdl-38466819

RÉSUMÉ

The effective delivery of synthetic RNA into mononuclear phagocytes is a prerequisite for experimental research and therapeutic development. However, traditional methods are highly ineffective and toxic for these cells. Here, we aimed to optimize a transfection protocol for primary bone marrow-derived phagocytes, specifically dendritic cells and macrophages, using lipid nanoparticles generated by microfluidics. Our results show that a lipid mixture similar to that used in Moderna's COVID-19 messenger RNA vaccine outperforms the others tested. Improved messenger RNA transfection can be achieved by replacing uridine with methylpseudouridine but not methoxyuridine, which interferes with transfection. The addition of diphenyleneiodonium or apocynin can enhance transfection in a cell type-dependent manner without adverse effects, while apolipoprotein E provides no added value. These optimized transfection conditions can also be used for microRNA agonists and antagonists. In sum, this study offers a straightforward, highly efficient, reproducible, and nontoxic protocol to deliver RNA into different primary mononuclear phagocytes in culture.


Sujet(s)
Nanoparticules , Transfection , Nanoparticules/composition chimique , Transfection/méthodes , Animaux , Cellules dendritiques , Phagocytes/métabolisme , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Lipides/composition chimique , Souris , Humains , ARN messager/génétique , microARN/génétique , Cellules cultivées , Souris de lignée C57BL , Liposomes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE