16 research outputs found

    Adipose tissue macrophage heterogeneity and the role of Tim4⁺ macrophages in lipid homeostasis

    Get PDF
    Resident macrophages are essential for the maintenance of tissue homeostasis as they participate in clearance of apoptotic cells and tissue remodelling and repair. In recent years, there has been an increased interest in the study of adipose tissue macrophages (ATMs). In lean individuals, ATMs are important for the control of insulin sensitivity, thermogenesis, angiogenesis and adipose tissue development. In obesity, the number and phenotype of ATMs is altered, and is associated with chronic low grade systemic and local inflammation. These “pro-inflammatory” changes are postulated to contribute to the manifestation of metabolic syndrome. These findings have suggested that the pool of ATMs is heterogeneous and may change, especially during obesity. To date, the characterisation of ATMs has been limited largely to the F4/80/CD11b markers, however the hypothesis of this thesis is that ATMs have distinct phenotype and function that could influence, in different ways, tissue homeostasis. This thesis aims to characterise and phenotype ATM subsets in order to better understand their potential specific role in the tissue. During the course of this research, a novel population of Tim4+ resident ATMs were identified. An additional aim of this thesis was to elucidate their role in adipose tissue homeostasis. Partial bone marrow chimeras were used to identify macrophage origin. The main AT depots were shielded from irradiation and a donor BM was injected intravenously. After 8 weeks, the origin of macrophages was analysed using flow cytometry. Tim4, a phosphatidylserine receptor mediating phagocytosis of apoptotic cells and a marker found on resident macrophages in other tissues, was used for the first time in adipose tissue. Four subsets of ATMs were identified: F4/80highCD11c-Tim4+, F4/80highCD11c- Tim4-; F4/80lowCD11c+Tim4-; F4/80lowCD11c-Tim4-. Interestingly, this newly described F4/80highTim4+ ATM subset showed the lowest non-host chimerism compared to the other ATMs, suggesting this is a main self-replenishing resident ATM population. To study the impact of obesity on ATM turnover, partial chimeric mice were fed HFD for 8 weeks. This increased the number of macrophages in AT. However, the different subsets of ATMs were differentially affected by the diet. Indeed, only a small proportion of Tim4+ ATMs derived from the bone marrow. In contrast, replenishment of the 3 other subsets was almost fully dependent on the arrival of monocyte-derived cells from the bone marrow. TIMD4, the gene encoding for Tim4, has been highlighted in genetic studies as being linked with dyslipidaemia. This suggests that Tim4+ ATMs might play a role in lipid homeostasis. Further characterisation of Tim4 ATMs demonstrated that these Tim4+ ATMs are highly charged in neutral lipid, and also have an increased lysosomal activity (shown by lysotracker staining) compared to the other ATM subsets. Using blocking anti-Tim4 antibodies in vivo, I found that Tim4 contributed markedly to free fatty acid (FFA) release into the plasma after short-term and long term HFD feeding. In addition, in vitro and in vivo experiments demonstrated that Tim4 could be required for the uptake of neutral lipids and their integration into lysosomes for degradation, though this seems to be dependent on the nature of the lipid. Collectively, these results indicate that Tim4 plays a crucial role in the control of lipid trafficking under conditions when dietary lipid is in excess. Tim4 allows uptake of lipids by Tim4+ ATMs and subsequent release of FFA into the circulation. Finally, the presence of Tim4+ lipid laden ATMs was demonstrated in the human omentum. This finding may lead to the discovery of new targets to improve metabolic health in obese patients. This work stresses the importance of resident ATM population in body lipid homeostasis as they could be involved in coping with lipid availability in the body and influence the amount of FFA in the plasma

    Induction of IL-4R alpha-dependent microRNAs identifies PI3K/Akt signaling as essential for IL-4-driven murine macrophage proliferation in vivo

    Get PDF
    Macrophage (MΦ) activation must be tightly controlled to preclude overzealous responses that cause self-damage. MicroRNAs promote classical MΦ activation by blocking antiinflammatory signals and transcription factors but also can prevent excessive TLR signaling. In contrast, the microRNA profile associated with alternatively activated MΦ and their role in regulating wound healing or antihelminthic responses has not been described. By using an in vivo model of alternative activation in which adult Brugia malayi nematodes are implanted surgically in the peritoneal cavity of mice, we identified differential expression of miR-125b-5p, miR-146a-5p, miR-199b-5p, and miR-378-3p in helminth-induced MΦ. In vitro experiments demonstrated that miR-378-3p was specifically induced by IL-4 and revealed the IL-4–receptor/PI3K/Akt-signaling pathway as a target. Chemical inhibition of this pathway showed that intact Akt signaling is an important enhancement factor for alternative activation in vitro and in vivo and is essential for IL-4–driven MΦ proliferation in vivo. Thus, identification of miR-378-3p as an IL-4Rα–induced microRNA led to the discovery that Akt regulates the newly discovered mechanism of IL-4–driven macrophage proliferation. Together, the data suggest that negative regulation of Akt signaling via microRNAs might play a central role in limiting MΦ expansion and alternative activation during type 2 inflammatory settings

    Macrophage origin limits functional plasticity in helminth-bacterial co-infection

    Get PDF
    Rapid reprogramming of the macrophage activation phenotype is considered important in the defense against consecutive infection with diverse infectious agents. However, in the setting of persistent, chronic infection the functional importance of macrophage-intrinsic adaptation to changing environments vs. recruitment of new macrophages remains unclear. Here we show that resident peritoneal macrophages expanded by infection with the nematode Heligmosomoides polygyrus bakeri altered their activation phenotype in response to infection with Salmonella enterica ser. Typhimurium in vitro and in vivo. The nematode-expanded resident F4/80high macrophages efficiently upregulated bacterial induced effector molecules (e.g. MHC-II, NOS2) similarly to newly recruited monocyte-derived macrophages. Nonetheless, recruitment of blood monocyte-derived macrophages to Salmonella infection occurred with equal magnitude in co-infected animals and caused displacement of the nematode-expanded, tissue resident-derived macrophages from the peritoneal cavity. Global gene expression analysis revealed that although nematode-expanded resident F4/80high macrophages made an anti-bacterial response, this was muted as compared to newly recruited F4/80low macrophages. However, the F4/80high macrophages adopted unique functional characteristics that included enhanced neutrophil-stimulating chemokine production. Thus, our data provide important evidence that plastic adaptation of MΦ activation does occur in vivo, but that cellular plasticity is outweighed by functional capabilities specific to the tissue origin of the cell

    IL-4 directly signals tissue-resident macrophages to proliferate beyond homeostatic levels controlled by CSF-1

    Get PDF
    Macrophages (M Phi s) colonize tissues during inflammation in two distinct ways: recruitment of monocyte precursors and proliferation of resident cells. We recently revealed a major role for IL-4 in the proliferative expansion of resident M Phi s during a Th2-biased tissue nematode infection. We now show that proliferation of M Phi s during intestinal as well as tissue nematode infection is restricted to sites of IL-4 production and requires M Phi-intrinsic IL-4R signaling. However, both IL-4R alpha-dependent and -independent mechanisms contributed to M Phi proliferation during nematode infections. IL-4R-independent proliferation was controlled by a rise in local CSF-1 levels, but IL-4R alpha expression conferred a competitive advantage with higher and more sustained proliferation and increased accumulation of IL-4R alpha(+) compared with IL-4R alpha(-) cells. Mechanistically, this occurred by conversion of IL-4R alpha(+) M Phi s from a CSF-1-dependent to -independent program of proliferation. Thus, IL-4 increases the relative density of tissue M Phi s by overcoming the constraints mediated by the availability of CSF-1. Finally, although both elevated CSF1R and IL-4R alpha signaling triggered proliferation above homeostatic levels, only CSF-1 led to the recruitment of monocytes and neutrophils. Thus, the IL-4 pathway of proliferation may have developed as an alternative to CSF-1 to increase resident M Phi numbers without coincident monocyte recruitment

    Ym1 induces RELMα and rescues IL-4Rα deficiency in lung repair during nematode infection

    Get PDF
    <div><p>Ym1 and RELMα are established effector molecules closely synonymous with Th2-type inflammation and associated pathology. Here, we show that whilst largely dependent on IL-4Rα signaling during a type 2 response, Ym1 and RELMα also have IL-4Rα-independent expression patterns in the lung. Notably, we found that Ym1 has opposing effects on type 2 immunity during nematode infection depending on whether it is expressed at the time of innate or adaptive responses. During the lung migratory stage of <i>Nippostrongylus brasiliensis</i>, Ym1 promoted the subsequent reparative type 2 response but once that response was established, IL-4Rα-dependent Ym1 was important for limiting the magnitude of type 2 cytokine production from both CD4+ T cells and innate lymphoid cells in the lung. Importantly, our study demonstrates that delivery of Ym1 to IL-4Rα deficient animals drives RELMα production and overcomes lung repair deficits in mice deficient in type 2 immunity. Together, Ym1 and RELMα, exhibit time and dose-dependent interactions that determines the outcome of lung repair during nematode infection.</p></div

    Fibre-based sensing for bacterial detection with time-resolved single photon spectroscopy

    No full text
    In the dawn of multiple drug resistant (MDR) infections, there is an unmet need for fast, accurate and less invasive diagnostic methods to identify lung infection and inflammation. One approach to tackle this challenge is by combining optical imaging agents with imaging or sensing through optical fibres. Previous approaches use different fibred imaging modalities - spectral, ratiometric, and time resolved. These techniques require a complex optical fibre bundle, and can be limited by distortions from this imaging fibre while also suffering due to tissue autofluorescence and photobleaching. Recently, complementary metal-oxide semiconductor (CMOS) single photon avalanche diode (SPAD) line sensors are becoming increasingly popular for applications such as time-resolved fluorescence sensing (TRFS). They are highly multiplexed, with good collection efficiency and time resolution (&lt; 500 ps time stamping resolution) which enables the recording of luminescent kinetics down to nanoseconds. The response from excited exogenous fluorophores can be investigated in three dimensions to enhance selectivity and sensitivity: fluorescent intensity, fluorescence emission spectral shape and fluorescence lifetime. We demonstrate that the use of fluorescence emission lifetime in combination with the spectral shape can confirm the presence of bacteria labelled with optical imaging agents, overcoming the known challenge of tissue autofluorescence. Variation in both parameters can also indicate changes in the morphology of bacteria introduced through treatments.Presented here is an endoscopic approach for optical fibre-based sensing of bacterial infections with clinical molecular probes for direct detection of gram-negative bacteria in size restricted regions such as the alveolar space of the distal lung. While this has been previously demonstrated in fibred imaging modalities, we demonstrate selectivity without reliance on spatial information offering faster interventions. Furthermore, time-resolved spectroscopy allows the use of off-the-shelf optical fibres and makes complex fibre probe development unnecessary, hence enabling deployment of miniaturised disposable fibre probes.<br/

    FluoroPi Device With SmartProbes: A Frugal Point-of-Care System for Fluorescent Detection of Bacteria From a Pre-Clinical Model of Microbial Keratitis.

    Get PDF
    PurposeRapid and accurate diagnosis of microbial keratitis (MK) could greatly improve patient outcomes. Here, we present the development of a rapid, accessible multicolour fluorescence imaging device (FluoroPi) and evaluate its performance in combination with fluorescent optical reporters (SmartProbes) to distinguish bacterial Gram status. Furthermore, we show feasibility by imaging samples obtained by corneal scrape and minimally invasive corneal impression membrane (CIM) from ex vivo porcine corneal MK models.MethodsFluoroPi was built using a Raspberry Pi single-board computer and camera, light-emitting-diodes (LEDs), and filters for white-light and fluorescent imaging, with excitation and detection of bacterial optical SmartProbes: Gram-negative, NBD-PMX (exmax 488 nm); Gram positive, Merocy-Van (exmax 590 nm). We evaluated FluoroPi with bacteria (Pseudomonas aeruginosa and Staphylococcus aureus) isolated from ex vivo porcine corneal models of MK by scrape (needle) and CIM with the SmartProbes.ResultsFluoroPi provides ConclusionsFluoroPi coupled with SmartProbes provides effective, low-cost bacterial imaging, delineating Gram-negative and Gram-positive bacteria directly sampled from a preclinical model of MK.Translational relevanceThis study provides a crucial stepping stone toward clinical translation of a rapid, minimally invasive diagnostic approach for MK

    Helminth-expanded MΦ upregulate NOS2 and MHC-II expression following bacterial infection.

    No full text
    <p>(A) Intracellular NOS2 staining in peritoneal MΦ from SL3261 (SL3261), <i>H</i>. <i>polygyrus</i> (H.poly) co-infected (coinf.) or naive mice. (B) Representative dot plots depicting NOS2 vs. F4/80 expression in peritoneal MΦ isolated in A. (C) Quantitative analysis of NOS2 expression in F4/80<sup>high</sup> (high) or F4/80<sup>low</sup> (low) MΦ isolated in A (D) Intracellular Relm-α staining in MΦ isolated in A. (E) Quantitative analysis of NOS2 expression in Relm-α positive (pos) or negative (neg) MΦ isolated in A (F) MHC-II expression (geometric mean fluorescence intensity) on F4/80<sup>high</sup> and F4/80<sup>low</sup> MΦ isolated in A. (G) Splenic bacterial burden in single SL3261 (SL3261) or consecutively <i>H</i>. <i>polygyrus</i>.<i>/</i> SL3261 co-infected (H.poly + SL3261) mice. Data pooled from 4 separate experiments with 5–6 animals per group per experiment. Datapoints depict individual animals and bars indicate mean and SEM. (H) As in G utilizing indicated infection doses of SL3261. One representative experiment of 2 with 5 animals per group shown. *: p<0.05; **: p<0.01, ***; p<0.001; n.s.: not significant.</p
    corecore