Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.829
1.
Front Immunol ; 15: 1383498, 2024.
Article En | MEDLINE | ID: mdl-38827743

This study investigates immune priming effects associated with granulocytes in crickets through a comprehensive analysis. Kaplan-Meier survival analysis reveals a significant contrast in survival rates, with the heat-killed Bacillus thuringiensis (Bt)-primed group exhibiting an impressive ~80% survival rate compared to the PBS buffer-primed group with only ~10% survival 60 hours post live Bt infection. Hemocyte analysis underscores elevated hemocyte counts, particularly in granulocytes of the killed Bt-primed group, suggesting a correlation between the heat-killed Bt priming and heightened immune activation. Microscopy techniques further explore granulocyte morphology, unveiling distinctive immune responses in the killed Bt-primed group characterized by prolonged immune activation, heightened granulocyte activity, phagocytosis, and extracellular trap formation, contributing to enhanced survival rates. In particular, after 24 hours of injecting live Bt, most granulocytes in the PBS buffer-primed group exhibited extracellular DNA trap cell death (ETosis), while in the killed Bt-primed group, the majority of granulocytes were observed to maintain highly activated extracellular traps, sustaining the immune response. Gene expression analysis supports these findings, revealing differential regulation of immune-related genes such as antibacterial humoral response, detection of bacterial lipopeptides, and cellular response to bacteria lipopeptides. Additionally, the heat-killed Bt-primed group, the heat-killed E. coli-primed group, and the PBS-primed group were re-injected with live Bt 2 and 9 days post priming. Two days later, only the PBS-primed group displayed low survival rates. After injecting live Bt 9 days later, the heat-killed E. coli-primed group surprisingly showed a similarly low survival rate, while the heat-killed Bt-primed group exhibited a high survival rate of ~60% after 60 hours, with actively moving and healthy crickets. In conclusion, this research provides valuable insights into both short-term and long-term immune priming effects in crickets, contributing to our understanding of invertebrate immunity with potential applications in public health.


Bacillus thuringiensis , Granulocytes , Gryllidae , Animals , Granulocytes/immunology , Gryllidae/immunology , Bacillus thuringiensis/immunology , Phagocytosis/immunology , Hemocytes/immunology , Extracellular Traps/immunology
2.
Front Immunol ; 15: 1415573, 2024.
Article En | MEDLINE | ID: mdl-38835772

Efferocytosis, the process of engulfing and removing apoptotic cells, plays an essential role in preserving tissue health and averting undue inflammation. While macrophages are primarily known for this task, dendritic cells (DCs) also play a significant role. This review delves into the unique contributions of various DC subsets to efferocytosis, highlighting the distinctions in how DCs and macrophages recognize and handle apoptotic cells. It further explores how efferocytosis influences DC maturation, thereby affecting immune tolerance. This underscores the pivotal role of DCs in orchestrating immune responses and sustaining immune equilibrium, providing new insights into their function in immune regulation.


Dendritic Cells , Macrophages , Phagocytosis , Dendritic Cells/immunology , Humans , Phagocytosis/immunology , Animals , Macrophages/immunology , Apoptosis/immunology , Immune Tolerance , Efferocytosis
3.
Sci Immunol ; 9(96): eadl2388, 2024 Jun 07.
Article En | MEDLINE | ID: mdl-38848343

Professional phagocytes like neutrophils and macrophages tightly control what they consume, how much they consume, and when they move after cargo uptake. We show that plasma membrane abundance is a key arbiter of these cellular behaviors. Neutrophils and macrophages lacking the G protein subunit Gß4 exhibited profound plasma membrane expansion, accompanied by marked reduction in plasma membrane tension. These biophysical changes promoted the phagocytosis of bacteria, fungus, apoptotic corpses, and cancer cells. We also found that Gß4-deficient neutrophils are defective in the normal inhibition of migration following cargo uptake. Sphingolipid synthesis played a central role in these phenotypes by driving plasma membrane accumulation in cells lacking Gß4. In Gß4 knockout mice, neutrophils not only exhibited enhanced phagocytosis of inhaled fungal conidia in the lung but also increased trafficking of engulfed pathogens to other organs. Together, these results reveal an unexpected, biophysical control mechanism central to myeloid functional decision-making.


Cell Membrane , Mice, Knockout , Phagocytosis , Animals , Phagocytosis/immunology , Cell Membrane/metabolism , Cell Membrane/immunology , Mice , Myeloid Cells/immunology , Mice, Inbred C57BL , Neutrophils/immunology , Macrophages/immunology
4.
Front Immunol ; 15: 1372904, 2024.
Article En | MEDLINE | ID: mdl-38742116

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.


Phagocytes , Phagocytosis , Recombinant Proteins , Animals , Phagocytosis/immunology , Phagocytes/immunology , Phagocytes/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Protein Binding , Strongylocentrotus purpuratus/immunology , Strongylocentrotus purpuratus/genetics , Immunity, Innate , Protein Isoforms/genetics , Protein Isoforms/immunology , Sea Urchins/immunology , Vibrio/immunology , Opsonin Proteins/metabolism , Opsonin Proteins/immunology
5.
J Immunol ; 212(12): 1932-1944, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38709167

IFN regulatory factor 7 (IRF7) exerts anti-infective effects by promoting the production of IFNs in various bacterial and viral infections, but its role in highly morbid and fatal Candida albicans infections is unknown. We unexpectedly found that Irf7 gene expression levels were significantly upregulated in tissues or cells after C. albicans infection in humans and mice and that IRF7 actually exacerbates C. albicans infection in mice independent of its classical function in inducing IFNs production. Compared to controls, Irf7-/- mice showed stronger phagocytosis of fungus, upregulation of C-type lectin receptor CD209 expression, and enhanced P53-AMPK-mTOR-mediated autophagic signaling in macrophages after C. albicans infection. The administration of the CD209-neutralizing Ab significantly hindered the phagocytosis of Irf7-/- mouse macrophages, whereas the inhibition of p53 or autophagy impaired the killing function of these macrophages. Thus, IRF7 exacerbates C. albicans infection by compromising the phagocytosis and killing capacity of macrophages via regulating CD209 expression and p53-AMPK-mTOR-mediated autophagy, respectively. This finding reveals a novel function of IRF7 independent of its canonical IFNs production and its unexpected role in enhancing fungal infections, thus providing more specific and effective targets for antifungal therapy.


Autophagy , Candida albicans , Candidiasis , Interferon Regulatory Factor-7 , Lectins, C-Type , Macrophages , Mice, Knockout , Phagocytosis , Receptors, Cell Surface , TOR Serine-Threonine Kinases , Animals , Mice , Phagocytosis/immunology , Autophagy/immunology , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Candidiasis/immunology , Candida albicans/immunology , Candida albicans/physiology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factor-7/immunology , Macrophages/immunology , Humans , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Mice, Inbred C57BL , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Signal Transduction/immunology
6.
Life Sci Alliance ; 7(8)2024 Aug.
Article En | MEDLINE | ID: mdl-38803222

The merozoite surface protein 1 (MSP1) is the most abundant protein on the surface of the invasive merozoite stages of Plasmodium falciparum and has long been considered a key target of protective immunity. We used samples from a single controlled human malaria challenge study to test whether the full-length version of MSP1 (MSP1FL) induced antibodies that mediated Fc-IgG functional activity in five independent assays. We found that anti-MSP1FL antibodies induced complement fixation via C1q, monocyte-mediated phagocytosis, neutrophil respiratory burst, and natural killer cell degranulation as well as IFNγ production. Activity in each of these assays was strongly associated with protection. The breadth of MSP1-specific Fc-mediated effector functions was more strongly associated with protection than the individual measures and closely mirrored what we have previously reported using the same assays against merozoites. Our findings suggest that MSP1FL is an important target of functional antibodies that contribute to a protective immune response against malaria.


Antibodies, Protozoan , Malaria, Falciparum , Merozoite Surface Protein 1 , Phagocytosis , Plasmodium falciparum , Humans , Merozoite Surface Protein 1/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium falciparum/immunology , Antibodies, Protozoan/immunology , Phagocytosis/immunology , Immunoglobulin G/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Interferon-gamma/metabolism , Interferon-gamma/immunology , Female , Merozoites/immunology , Neutrophils/immunology , Neutrophils/metabolism
7.
Clin Immunol ; 263: 110206, 2024 Jun.
Article En | MEDLINE | ID: mdl-38599263

Patients suffering from sepsis-induced acute lung injury (ALI) exhibit a high mortality rate, and their prognosis is closely associated with infiltration of neutrophils into the lungs. In this study, we found a significant elevation of CD64+ neutrophils, which highly expressed p75 neurotrophin receptor (p75NTR) in peripheral blood of mice and patients with sepsis-induced ALI. p75NTR+CD64+ neutrophils were also abundantly expressed in the lung of ALI mice induced by lipopolysaccharide. Conditional knock-out of the myeloid lineage's p75NTR gene improved the survival rates, attenuated lung tissue inflammation, reduced neutrophil infiltration and enhanced the phagocytic functions of CD64+ neutrophils. In vitro, p75NTR+CD64+ neutrophils exhibited an upregulation and compromised phagocytic activity in blood samples of ALI patients. Blocking p75NTR activity by soluble p75NTR extracellular domain peptide (p75ECD-Fc) boosted CD64+ neutrophils phagocytic activity and reduced inflammatory cytokine production via regulation of the NF-κB activity. The findings strongly indicate that p75NTR+CD64+ neutrophils are a novel pathogenic neutrophil subpopulation promoting sepsis-induced ALI.


Acute Lung Injury , Mice, Inbred C57BL , Neutrophils , Phagocytosis , Receptors, IgG , Receptors, Nerve Growth Factor , Sepsis , Animals , Acute Lung Injury/immunology , Acute Lung Injury/etiology , Neutrophils/immunology , Neutrophils/metabolism , Sepsis/immunology , Sepsis/complications , Humans , Receptors, IgG/metabolism , Receptors, IgG/genetics , Receptors, IgG/immunology , Mice , Male , Phagocytosis/immunology , Receptors, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/immunology , Mice, Knockout , Lipopolysaccharides , Cytokines/metabolism , Cytokines/immunology , Lung/immunology , Lung/pathology , Female , NF-kappa B/metabolism , NF-kappa B/immunology , Nerve Tissue Proteins
8.
Trends Immunol ; 45(5): 322-324, 2024 May.
Article En | MEDLINE | ID: mdl-38644134

Interferons and central nervous system resident macrophages, microglia, are well-known for their respective roles in antiviral defense and phagocytosis. Using a classic experimental paradigm for examining activity-dependent neural plasticity, Escoubas, Dorman, et al. recently identified a role for microglial type I interferon signaling in the clearance of unwanted neurons during mouse brain development.


Brain , Interferon Type I , Microglia , Animals , Brain/immunology , Brain/growth & development , Interferon Type I/metabolism , Interferon Type I/immunology , Mice , Microglia/immunology , Microglia/metabolism , Humans , Signal Transduction/immunology , Neurons/immunology , Neurons/metabolism , Phagocytosis/immunology , Neuronal Plasticity/immunology
9.
Science ; 384(6691): eabo7027, 2024 Apr 05.
Article En | MEDLINE | ID: mdl-38574142

Macrophages are functionally heterogeneous cells essential for apoptotic cell clearance. Apoptotic cells are defined by homogeneous characteristics, ignoring their original cell lineage identity. We found that in an interleukin-4 (IL-4)-enriched environment, the sensing of apoptotic neutrophils by macrophages triggered their tissue remodeling signature. Engulfment of apoptotic hepatocytes promoted a tolerogenic phenotype, whereas phagocytosis of T cells had little effect on IL-4-induced gene expression. In a mouse model of parasite-induced pathology, the transfer of macrophages conditioned with IL-4 and apoptotic neutrophils promoted parasitic egg clearance. Knockout of phagocytic receptors required for the uptake of apoptotic neutrophils and partially T cells, but not hepatocytes, exacerbated helminth infection. These findings suggest that the identity of apoptotic cells may contribute to the development of distinct IL-4-driven immune programs in macrophages.


Apoptosis , Interleukin-4 , Macrophages , Phagocytosis , Schistosomiasis mansoni , Animals , Mice , Apoptosis/immunology , Hepatocytes/immunology , Interleukin-4/genetics , Interleukin-4/metabolism , Macrophages/immunology , Mice, Knockout , Neutrophils/immunology , Phagocytosis/immunology , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/immunology , Disease Models, Animal
10.
J Autoimmun ; 145: 103196, 2024 May.
Article En | MEDLINE | ID: mdl-38458075

Type 1 diabetes (T1D) results from a breakdown in immunological tolerance, with pivotal involvement of antigen-presenting cells. In this context, antigen-specific immunotherapies have been developed to arrest autoimmunity, such as phosphatidylserine (PS)-liposomes. However, the role of certain antigen-presenting cells in immunotherapy, particularly human macrophages (Mφ) in T1D remains elusive. The aim of this study was to determine the role of Mφ in antigen-specific immune tolerance and T1D. To that end, we evaluated Mφ ability to capture apoptotic-body mimicking PS-liposomes in mice and conducted a phenotypic and functional characterisation of four human monocyte-derived Mφ (MoMφ) subpopulations (M0, M1, M2a and M2c) after PS-liposomes uptake. Our findings in mice identified Mφ as the most phagocytic cell subset in the spleen and liver. In humans, while phagocytosis rates were comparable between T1D and control individuals, PS-liposome capture dynamics differed among Mφ subtypes, favouring inflammatory (M1) and deactivated (M2c) Mφ. Notably, high nanoparticle concentrations did not affect macrophage viability. PS-liposome uptake by Mφ induced alterations in membrane molecule expression related to immunoregulation, reduced secretion of IL-6 and IL-12, and diminished autologous T-cell proliferation in the context of autoantigen stimulation. These results underscore the tolerogenic effects of PS-liposomes and emphasize their potential to target human Mφ, providing valuable insights into the mechanism of action of this preclinical immunotherapy.


Autoantigens , Diabetes Mellitus, Type 1 , Immunotherapy , Liposomes , Macrophages , Phosphatidylserines , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 1/immunology , Animals , Humans , Phosphatidylserines/metabolism , Phosphatidylserines/immunology , Mice , Immunotherapy/methods , Macrophages/immunology , Macrophages/metabolism , Autoantigens/immunology , Female , Immune Tolerance , Phagocytosis/immunology , Male , Mice, Inbred NOD , Autoimmunity , Adult
11.
J Autoimmun ; 145: 103197, 2024 May.
Article En | MEDLINE | ID: mdl-38447248

BACKGROUND AND OBJECTIVE: Understanding the regulation of efferocytosis by myeloid phagocytes is important in identifying novel targets in systemic lupus erythematosus (SLE). Cadherin-11 (CDH11), a cell adhesion molecule, is implicated in inflammatory arthritis and fibrosis and recently been shown to regulate macrophage phagocytosis. The extent and mechanism of this regulation is unknown. Our objective was to examine the extent to which CDH11 regulates myeloid phagocytes and contributes to autoimmunity and tissue inflammation. METHODS: We analyzed efferocytosis in macrophages and dendritic cells (DCs) from WT and Cdh11-/- mice and investigated the mechanisms in vitro. We investigated the role of CDH11 in disease development in vivo using the pristane induced lupus model. To translate the clinical relevance of CDH11 in human disease, we measured serum CDH11 levels in two independent pediatric SLE (pSLE) cohorts and healthy controls. RESULTS: Using bone marrow derived macrophages (BMDMs) and DCs (BMDCs), we found impaired efferocytosis in phagocytes from Cdh11-/- mice, mediated by downregulated efferocytosis receptor expression and RhoGTPase activation. Specifically, loss of CDH11 downregulated Mertk expression and Rac1 activation in BMDMs, and integrin αVß3 expression and Cdc42 activation in BMDCs, highlighting distinct pathways. In vivo, Cdh11-/- mice displayed defective efferocytosis and increased accumulation of apoptotic debris in pristane-induced lupus. Further, Cdh11-/- mice had enhanced systemic inflammation and autoimmune inflammation with increased anti-dsDNA autoantibodies, splenomegaly, type I interferons, and inflammatory cytokines. Paradoxically, at the tissue level, Cdh11-/- mice were protected against glomerulonephritis, indicating a dual role in murine lupus. Finally, SLE patients had increased serum CDH11 compared to controls. CONCLUSION: This study highlights a novel role of CDH11 in regulating myeloid cells and efferocytosis and its potential as a contributor to development in autoimmunity murine lupus. Despite the increase in autoimmunity, Cdh11-/- mice developed decreased tissue inflammation and damage.


Cadherins , Dendritic Cells , Disease Models, Animal , Lupus Erythematosus, Systemic , Macrophages , Phagocytosis , Animals , Child , Female , Humans , Mice , Autoimmunity , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/metabolism , Cadherins/metabolism , Cadherins/genetics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Inflammation/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/genetics , Macrophages/immunology , Macrophages/metabolism , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Phagocytosis/immunology , Terpenes
12.
Eur J Immunol ; 54(6): e2350771, 2024 Jun.
Article En | MEDLINE | ID: mdl-38494423

Vomocytosis, also known as nonlytic exocytosis, is a process whereby fully phagocytosed microbes are expelled from phagocytes without discernible damage to either the phagocyte or microbe. Although this phenomenon was first described in the opportunistic fungal pathogen Cryptococcus neoformans in 2006, to date, mechanistic studies have been hampered by an inability to reliably stimulate or inhibit vomocytosis. Here we present the fortuitous discovery that macrophages lacking the scavenger receptor MAcrophage Receptor with COllagenous domain (MARCO), exhibit near-total vomocytosis of internalised cryptococci within a few hours of infection. Marco-/- macrophages also showed elevated vomocytosis of a yeast-locked C. albicans strain, suggesting this to be a broadly relevant observation. We go on to show that MARCO's role in modulating vomocytosis is independent of its role as a phagocytic receptor, suggesting that this protein may play an important and hitherto unrecognised role in modulating macrophage behaviour.


Cryptococcus neoformans , Macrophages , Receptors, Immunologic , Animals , Mice , Cryptococcus neoformans/immunology , Macrophages/immunology , Macrophages/microbiology , Receptors, Immunologic/metabolism , Receptors, Immunologic/immunology , Receptors, Immunologic/genetics , Candida albicans/immunology , Phagocytosis/immunology , Mice, Knockout , Exocytosis/immunology , Cryptococcosis/immunology
13.
Exp Mol Med ; 55(1): 215-227, 2023 01.
Article En | MEDLINE | ID: mdl-36635431

Conflicting results on melatonin synthesis in multiple sclerosis (MS) have been reported due to variabilities in patient lifestyles, which are not considered when supplementing melatonin. Since melatonin acts through its receptors, we identified melatonin receptors in oligodendrocytes (OLs) in the corpus callosum, where demyelination occurs; the subventricular zone, where neural stem/progenitor cells (NSPCs) are located; and the choroid plexus, which functions as a blood-cerebrospinal fluid barrier. Moreover, using chimeric mice, resident macrophages were found to express melatonin receptors, whereas bone marrow-derived macrophages lost this expression in the demyelinated brain. Next, we showed that cuprizone-fed mice, which is an MS model, tended to have increased melatonin levels. While we used different approaches to alter the circadian rhythm of melatonin and cortisol, only the constant light approach increased NSPC proliferation and differentiation to oligodendrocyte precursor cells (OPCs), OPCs maturation to OLs and recruitment to the site of demyelination, the number of patrolling monocytes, and phagocytosis. In contrast, constant darkness and exogenous melatonin exacerbated these events and amplified monocyte infiltration. Therefore, melatonin should not be considered a universal remedy, as is currently claimed. Our data emphasize the importance of monitoring melatonin/cortisol oscillations in each MS patient by considering diet and lifestyle to avoid melatonin overdose.


Demyelinating Diseases , Melatonin , Monocytes , Multiple Sclerosis , Myelin Sheath , Phagocytosis , Animals , Mice , Cell Differentiation , Demyelinating Diseases/immunology , Demyelinating Diseases/metabolism , Disease Models, Animal , Hydrocortisone , Melatonin/pharmacology , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Phagocytosis/immunology , Receptors, Melatonin , Myelin Sheath/metabolism
14.
Front Immunol ; 13: 762080, 2022.
Article En | MEDLINE | ID: mdl-35145518

Leishmania spp. infection outcomes are dependent on both host and parasite factors. Manipulation of host signaling pathways involved in the generation of immune responses is thought to be one of the most common mechanisms used by parasites for persistence within the host. Considering the diversity of pathologies caused by different Leishmania spp., it is plausible that significant differences may exist in the mechanisms of host cell manipulation by each parasite species, which may have implications when developing new vaccine or treatment strategies. Here we show that in L. braziliensis-infection in BALB/c mice, a model of resistance, activation of ERK1/2 coincides with the peak of inflammatory responses and resolution of tissue parasitism. In contrast, in the susceptibility model of L. amazonensis-infection, an early silent phase of infection is observed, detected solely by quantification of parasite loads. At this early stage, only basal levels of P-ERK1/2 are observed. Later, after a brief shutdown of ERK1/2 phosphorylation, disease progression is observed and is associated with increased inflammation, lesion size and tissue parasitism. Moreover, the short-term down-regulation of ERK1/2 activation affected significantly downstream inflammatory pathways and adaptive T cell responses. Administration of U0126, a MEK/ERK inhibitor, confirmed this phenomenon, since bigger lesions and higher parasite loads were seen in infected mice that received U0126. To investigate how kinetics of ERK1/2 activation could affect the disease progression, U0126 was administered to L. amazonensis-infected animals earlier than the P-ERK1/2 switch off time-point. This intervention resulted in anticipation of the same effects on inflammatory responses and susceptibility phenotype seen in the natural course of infection. Additionally, in vitro inhibition of ERK1/2 affected the phagocytosis of L. amazonensis by BMDMs. Collectively, our findings reveal distinct temporal patterns of activation of inflammatory responses in L. braziliensis and L. amazonensis in the same animal background and a pivotal role for a brief and specific shutdown of ERK1/2 activation at late stages of L. amazonensis infection. Since activation of inflammatory responses is a crucial aspect for the control of infectious processes, these findings may be important for the search of new and specific strategies of vaccines and treatment for tegumentary leishmaniasis.


Immunity, Cellular , Leishmania mexicana/immunology , Leishmaniasis/immunology , Leishmaniasis/metabolism , Leishmaniasis/parasitology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Animals , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Female , Host-Pathogen Interactions/immunology , Inflammation Mediators/metabolism , Leishmaniasis/pathology , Mice , Parasite Load , Phagocytosis/immunology , Phosphorylation , Signal Transduction
15.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article En | MEDLINE | ID: mdl-35110413

Changes in population density lead to phenotypic differentiation of solitary and gregarious locusts, which display different resistance to fungal pathogens; however, how to regulate their cellular immune strategies remains unknown. Here, our stochastic simulation of pathogen proliferation suggested that humoral defense always enhanced resistance to fungal pathogens, while phagocytosis sometimes reduced defense against pathogens. Further experimental data proved that gregarious locusts had significantly decreased phagocytosis of hemocytes compared to solitary locusts. Additionally, transcriptional analysis showed that gregarious locusts promoted immune effector expression (gnbp1 and dfp) and reduced phagocytic gene expression (eater) and the cytokine tumor necrosis factor (TNF). Interestingly, higher expression of the cytokine TNF in solitary locusts simultaneously promoted eater expression and inhibited gnbp1 and dfp expression. Moreover, inhibition of TNF increased the survival of solitary locusts, and injection of TNF decreased the survival of gregarious locusts after fungal infection. Therefore, our results indicate that the alerted expression of TNF regulated the immune strategy of locusts to adapt to environmental changes.


Grasshoppers/immunology , Grasshoppers/microbiology , Immunity, Cellular/immunology , Metarhizium/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Gene Expression/immunology , Phagocytosis/immunology , Population Density , Transcription, Genetic/immunology
16.
Nat Commun ; 13(1): 662, 2022 02 03.
Article En | MEDLINE | ID: mdl-35115533

Increasingly, antibodies are being used to treat and prevent viral infections. In the context of HIV, efficacy is primarily attributed to dose-dependent neutralization potency and to a lesser extent Fc-mediated effector functions. It remains unclear whether augmenting effector functions of broadly neutralizing antibodies (bNAbs) may improve their clinical potential. Here, we use bNAb 10E8v4 targeting the membrane external proximal region (MPER) to examine the role of antibody-mediated effector and complement (C') activity when administered prophylactically against SHIV challenge in rhesus macaques. With sub-protective dosing, we find a 78-88% reduction in post-acute viremia that is associated with 10E8v4-mediated phagocytosis acting at the time of challenge. Neither plasma nor tissue viremic outcomes in vivo is improved with an Fc-modified variant of 10E8v4 enhanced for C' functions as determined in vitro. These results suggest that effector functions inherent to unmodified 10E8v4 contribute to efficacy against SHIVSF162P3 in the absence of plasma neutralizing titers, while C' functions are dispensable in this setting, informing design of bNAb modifications for improving protective efficacy.


Broadly Neutralizing Antibodies/immunology , Complement System Proteins/immunology , HIV Antibodies/immunology , HIV-1/immunology , Phagocytosis/immunology , Viremia/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Broadly Neutralizing Antibodies/metabolism , Broadly Neutralizing Antibodies/pharmacology , Cell Line, Tumor , Complement System Proteins/metabolism , Cytokines/immunology , Cytokines/metabolism , Female , HIV Antibodies/metabolism , HIV Antibodies/pharmacology , HIV Infections/immunology , HIV Infections/prevention & control , HIV Infections/virology , HIV-1/drug effects , HIV-1/physiology , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Macaca mulatta , Male , Phagocytosis/drug effects , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , Viremia/blood , Viremia/prevention & control
17.
Tuberculosis (Edinb) ; 132: 102165, 2022 01.
Article En | MEDLINE | ID: mdl-35045376

Mycobacterium tuberculosis (MTB) and M. avium-intracellulare complex (MAC) enter host phagocytes, such as neutrophils through lipoarabinomannan (LAM) binding to pattern-recognition receptors, inducing innate immune responses including phagocytosis. Phagocytosis of mycobacteria by human neutrophils depends on the binding of α(1 â†’ 2)-monomannose branching α(1 â†’ 6)-mannan core of LAM/lipomannan (LM), a common component among mycobacterial species, to lactosylceramide (LacCer)-enriched lipid microdomains. We investigated the binding specificities of several anti-LAM antibodies (Abs) to LAMs/LM and found anti-LAM monoclonal IgMs TMDU3 and LA066 were directed against mannan core. Each IgM showed different binding specificity to mannan core. Confocal and stimulated emission depletion microscopy revealed TMDU3 and LA066 strongly bind to MTB and MAC, respectively. Flow cytometric analysis revealed human neutrophils do not express Dectin-2, DC-SIGN or mannose receptor. Furthermore, neutrophil phagocytosis of mycobacteria was markedly inhibited by TMDU3 and LA066, respectively. Similarly, treatment of each mAb with neutrophils reduced the numbers of intracellular MAC. Together, our results suggest that the interaction of LacCer-enriched lipid microdomains with mannan core and its blocking are therapeutic or diagnostic targets for both TB and non-tuberculous mycobacteria infection.


Lipopolysaccharides/immunology , Mannans/metabolism , Mycobacterium/metabolism , Neutrophils/immunology , Phagocytosis/immunology , Adult , Humans , Lipopolysaccharides/analysis , Male , Middle Aged , Mycobacterium/immunology , Neutrophils/metabolism , Phagocytosis/genetics
18.
Sci Immunol ; 7(67): eabj5501, 2022 01 21.
Article En | MEDLINE | ID: mdl-35061505

The interaction of SIRPα with CD47 represents a major mechanism for preventing macrophage phagocytosis. However, CD47-independent mechanisms are poorly defined. Here, we report a critical role of SLAM family receptors (SFRs), ubiquitously expressed on hematopoietic cells and forming homotypic interactions, in constraining macrophage phagocytosis. We found that SFR deficiency triggered macrophage phagocytosis of hematopoietic cells, leading to severe rejection of donor hematopoietic graft in recipient mice. Specific SFR members, mainly SLAMF3 and SLAMF4, were identified as "don't eat me" receptors on macrophages. These receptors inhibited "eat me" signals, such as LRP1-mediated activation of mTOR and Syk, through SH2 domain-containing phosphatases. SFRs combined with, but were independent of, CD47 to mitigate macrophage phagocytosis, and the combined deletion of SFRs and CD47 resulted in hematopoietic cytopenia in mice. This SFR-mediated tolerance was compromised in patients with hemophagocytic lymphohistiocytosis, a syndrome characterized by inappropriate phagocytosis toward hematopoietic cells. Loss of SFRs potently elicited macrophage rejection of hematopoietic tumors. Deletion of SFRs also significantly enhanced the phagocytosis of CD19-positive hematopoietic targets by the macrophages expressing the chimeric CD19 antigen receptor. Therefore, SFR-mediated inhibition of macrophage phagocytosis is critical to hematopoietic homeostasis, and SFRs may represent previously unknown targets for tumor immunotherapy.


Hematologic Neoplasms/immunology , Immune Checkpoint Proteins/immunology , Macrophages/immunology , Phagocytosis/immunology , Signaling Lymphocytic Activation Molecule Family/immunology , Animals , Cell Line , Hematologic Neoplasms/pathology , Humans , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
19.
Dev Comp Immunol ; 129: 104348, 2022 04.
Article En | MEDLINE | ID: mdl-35026231

Matrix metalloproteinases (MMPs) are highly expressed in leukocytes and macrophages, which play a role in the innate immune response. Here, the cDNA sequence of MMP25 from Japanese sea bass (Lateolabrax japonicus) (LjMMP25) was identified. Phylogenetic analysis revealed that LjMMP25 was most closely related to large yellow croaker MMP25. Multiple sequence alignment of LjMMP25 with MMP25 sequences from other teleosts revealed that regions of known functional importance were highly conserved. Expression analysis revealed that LjMMP25 was highly expressed in the head kidney and widely expressed in other tissues including gill, spleen, and liver. LjMMP25 was found to regulate inflammatory cytokine production and promote phagocytosis and bacterial killing in monocytes/macrophages (MO/MФ). Furthermore, LjMMP25 regulated the inflammatory response by modulating NF-κB signaling. These findings reveal new information about the role of LjMMP25 in regulating pro-inflammatory responses in this species.


Bass/genetics , Amino Acid Sequence , Animals , Fish Diseases/immunology , Fish Proteins/genetics , GPI-Linked Proteins , Immunity, Innate/genetics , Leukocytes , Liver , Macrophages/immunology , Matrix Metalloproteinases, Membrane-Associated , Monocytes/immunology , Phagocytosis/immunology , Phylogeny , Sequence Alignment , Vibrio Infections/immunology
20.
Cell Rep ; 38(1): 110183, 2022 01 04.
Article En | MEDLINE | ID: mdl-34986357

Candida albicans is a commensal of the urogenital tract and the predominant cause of vulvovaginal candidiasis (VVC). Factors that increase circulatory estrogen levels such as pregnancy, the use of oral contraceptives, and hormone replacement therapy predispose women to VVC, but the reasons for this are largely unknown. Here, we investigate how adaptation of C. albicans to estrogen impacts the fungal host-pathogen interaction. Estrogen promotes fungal virulence by enabling C. albicans to avoid the actions of the innate immune system. Estrogen-induced innate immune evasion is mediated via inhibition of opsonophagocytosis through enhanced acquisition of the human complement regulatory protein, Factor H, on the fungal cell surface. Estrogen-induced accumulation of Factor H is dependent on the fungal cell surface protein Gpd2. The discovery of this hormone-sensing pathway might pave the way in explaining gender biases associated with fungal infections and may provide an alternative approach to improving women's health.


Candida albicans/immunology , Candidiasis, Vulvovaginal/pathology , Complement Pathway, Alternative/immunology , Estrogens/metabolism , Immune Evasion/immunology , Phagocytosis/immunology , Candida albicans/pathogenicity , Complement Factor H/metabolism , Female , Glycerol-3-Phosphate Dehydrogenase (NAD+)/metabolism , Humans , Immunity, Innate/immunology , Progesterone/metabolism , Virulence/immunology
...