Abstract
Professional phagocytes (such as macrophages1,2) and non-professional phagocytes3–9 (such as epithelial cells) clear billions of apoptotic cells and particles on a daily basis10,11. Since these phagocytes reside in proximity in most tissues, whether cross-communication exists between them during cell clearance, and how this might impact inflammation are not known12. Here, we show that macrophages, via the release of a soluble growth factor and microvesicles, redirect the type of particles engulfed by non-professional phagocytes and influence their inflammatory response. During apoptotic cell engulfment or in response to inflammation-associated cytokines, macrophages released insulin-like growth factor 1 (IGF-1). The binding of IGF-1 to its receptor on non-professional phagocytes redirected their phagocytosis, such that uptake of larger apoptotic cells was dampened while engulfment of microvesicles was enhanced. Macrophages were refractory to this IGF-1 mediated engulfment modulation. Macrophages also released microvesicles, whose uptake by epithelial cells, enhanced by IGF-1, led to decreased inflammatory responses by epithelial cells. Consistent with these observations, deletion of IGF-1 receptor in airway epithelial cells led to exacerbated lung inflammation after allergen exposure. These genetic and functional studies reveal a novel IGF-1 and microvesicle-dependent communication between macrophages and epithelial cells that can critically influence the magnitude of tissue inflammation in vivo.
To address potential cross-regulation among phagocytes, we initially tested a panel of eleven soluble mediators that have been linked to tissue repair, inflammation dampening, and tissue morphogenesis13, for their ability to modulate engulfment by LR73 fibroblasts, a non-professional phagocytic cell line. Of the eleven factors tested, only insulin-like growth factor 1 (IGF-1) significantly dampened apoptotic cell uptake, at concentrations reported in mouse and human serum (100–600 ng/mL)14 (Fig. 1a, 1b). Other factors such as EGF, FGF2, VEGF, PDGF-AA and PDGF-BB did not alter engulfment even over a range of concentrations (Extended Data Fig. 1a), although they all elicited early downstream signaling events (Extended Data Fig. 1b–e). The engulfment dampening effect of IGF-1 was also seen with airway epithelial cell lines BEAS-2B and 16HBE14o-, and the endothelial cell line SVEC-40 (Fig. 1c, Extended Data Fig. 1f, g).
The IGF-1 effect was not due to masking phosphatidylserine (PtdSer) on the apoptotic cells (Fig. 1d). LR73 cells express the IGF-1 receptor (IGF-1R) and IGF-1 treatment elicited phosphorylation of Akt, a signaling molecule downstream of IGF-1R (Fig. 1b). In blood, IGF-1 is bound to IGF binding proteins (IGFBPs) that stabilize and sequester IGF-115. Addition of IGFBP3 with IGF-1 restored the phagocytic capability of LR73 cells (Fig. 1e), suggesting that binding of active IGF-1 to IGF-1R was necessary. A neutralizing antibody against the human IGF-1R reversed the engulfment-dampening effect of IGF-1 on BEAS-2B cells (Fig. 1f). Further, OSI-906 (a small molecule kinase inhibitor of IGF-1R currently tested in clinical trials16), and another inhibitor, NVP-AEW541, rescued the engulfment capacity of LR73 cells treated with IGF-1, with concomitant decrease in phosphorylation of IGF-1R and Erk (Fig. 1g and Extended Data Fig. 2a)17. IGF-II and insulin, which share structural similarity with IGF-1 but have lower affinities to IGF-1R18, could also reduce apoptotic cell uptake (Extended Data Fig. 4), albeit at higher concentrations. Thus, productive signaling through the IGF-1R is necessary for IGF-1 to dampen apoptotic cell uptake.
In contrast to the inhibitory effect on apoptotic cell uptake, IGF-1 enhanced the uptake of PtdSer containing liposomes of 150–200nm in size (Fig. 1h). This was not seen with EGF or VEGF. IGF-1 enhancement required IGF-1R signaling, as the inhibitor OSI-906 reduced the increased liposome uptake (Fig. 1h and 1i). Thus, IGF-1 can redirect phagocytosis by non-professional phagocytes, suppressing uptake of larger apoptotic cells while enhancing internalization of smaller particles.
The IGF-1 effect was reversible, as washing phagocytes pre-treated with IGF-1 resulted in near complete restoration of engulfment (Fig. 1j). The effect of IGF-1 on phagocytes was also rapid, as adding IGF-1 simultaneously with apoptotic cells inhibited engulfment similar to pre-treated cells (data not shown), suggesting interference at early step(s) during phagocytosis. Blocking some early signaling pathways downstream of IGF-1R, such as Akt, mTOR, Erk or PI3-kinase19, did not rescue of the IGF-1 effect (Extended Data Fig. 3a–d). Although IGF-1 can activate RhoA, which is known to dampen apoptotic cell engulfment20, inhibiting RhoA-mediated signaling did not reverse IGF-1-mediated engulfment suppression (Extended Data Fig. 3e,f). Overexpressing activated Rac1 (which promotes apoptotic cell engulfment21,22) bypassed attenuation of apoptotic cell uptake (Fig. 1k), suggesting that IGF-1R acts at or before a step regulated by active Rac1. Since apoptotic cell uptake requires both Rac1-dependent actin polymerization and de-polymerization23, we explored actin regulation via IGF-1. Cytochalasin D (cytoD) that promotes actin depolymerization24. Phagocytosis of apoptotic cells was potently inhibited by cytoD4 (data not shown). While cytoD did not affect the basal uptake of liposomes by LR73 cells, it blocked the IGF-1 induced increase in liposome uptake (Fig. 1l). Of note, cytoD treated LR73 cells appeared morphologically normal for the duration of the assay. Latrunculin A, which promotes actin depolymerization via a different mechanism25, also reversed IGF-1 mediated enhancement of liposome uptake, without affecting the basal uptake of liposomes (Fig. 1m). Additionally, Arp2/3 complex can regulate phagocytosis through the formation of branched actin networks; however, CK-666, a small molecule inhibitor of Arp2/326, had minimal effect on the IGF-1 mediated increase of liposome uptake (Extended Data Fig. 3g). Collectively, IGF-1 mediated modulation of phagocytosis involves rapid and reversible modification of F-actin/G-actin dynamics, but likely not Arp2/3 mediated functions.
We next tested the effect of IGF-1 on professional phagocytes. Adding IGF-1 to the macrophage cell lines J774 or IC-21, or primary macrophages (bone marrow-derived or resident peritoneal macrophages), did not affect corpse uptake, even with supra-physiological concentrations of IGF-1, at every time point analyzed (Fig. 1n–p, and Extended Data Fig. 4a, d). Peritoneal macrophages exposed to IGF-1 also had comparable liposome uptake as control treated cells (Fig. 1q). Importantly, IGF-1R expression was confirmed in all macrophage cell types analyzed (Extended Data Fig. 4b) and IGF-1 initiated early signaling in macrophages, as assessed by IGF-1R and Akt phosphorylation (Fig. 1n, Extended Data Fig. 4a). These data suggest that IGF-1 mediated modulation of phagocytosis does not extend to macrophages.
High levels of serum IGF-1 is linked to IGF-1 production in the liver, but macrophages can also produce IGF-1 in response to the cytokine IL-427,28, raising the possibility that within a tissue, IGF-1 release from macrophages could regulate nearby non-professional phagocytes. We first confirmed IGF-1 secretion by peritoneal macrophages treated with IL-4 (Fig. 2a). Furthermore, resident peritoneal macrophages exposed to apoptotic Jurkat cells (but not live cells) produced IGF-1 (Fig. 2a). IGF-1 protein induction appears to be from newly transcribed Igf1 message (Extended Data Fig. 5a). Macrophage-produced IGF-1 could also suppress apoptotic cell uptake by LR73 cells, reversed by IGFBP3 addition (Fig. 2b).
To test the communication between phagocytes in vivo, we chose the lung, as alveolar macrophages and airway epithelial cells reside in close proximity, and both engulf apoptotic cells4. Alveolar macrophages, similar to the resident peritoneal macrophages, are derived from fetal monocytes29, and are readily isolated via CD11c+ Siglec F+ expression, while the airway epithelial cells can be isolated and tracked via available genetic tools30. IGF-1R expression was prominent in the airway epithelial cells (Extended Data Fig. 5b) but was also detectable at lower levels in alveolar macrophages. To test the effect of IGF-1 in vivo, we used mice where airway epithelial cells are specifically marked by YFP. When either apoptotic cells or liposomes were administered intranasally with IGF-1 (Fig. 2c), YFP+ airway epithelial cells had decreased apoptotic cell engulfment, but enhanced liposome uptake (Fig. 2d, e). Importantly, lung macrophages from the same IGF-1 treated mice were unaffected in their ability to engulf these targets (Fig. 2d, e). Thus, IGF-1 can differentially affect engulfment by professional and non-professional phagocytes closely residing in the same tissue.
We next tested IGF-1 release by alveolar macrophages in vivo. Intranasal administration of IL-4 or apoptotic Jurkat cells resulted in an increased level of IGF-1 in the bronchoalveolar lavage (BAL) fluid (Fig. 2f). When we tested IL-13 or IL-5 (cytokines linked to lung homeostasis and inflammation31), IL-13 (whose receptor shares a common subunit with the IL-4R32), but not IL-5, induced IGF-1 production (Fig. 2f). Since the source of IGF-1 is difficult to distinguish in these experiments, we generated LysM-Cre/Igf1fl/fl mice 33,34, targeting IGF-1 deletion in the myeloid lineage. LysM-Cre/Igf1fl/fl mice showed loss of Igf1 mRNA in alveolar macrophages (Extended Data Fig. 5c) and IGF-1 induction after IL-4, IL-13 or apoptotic cell treatment (Fig. 2H). This suggested the macrophage/myeloid population as the predominant source of inducible IGF-1 in the lung.
To investigate IGF-1/IGF-1R signaling in inflammation, we used a model of airway inflammation induced by the allergen house dust mite (HDM), where apoptotic cell recognition by airway epithelial cells influences inflammation4,35. We administered HDM intranasally in the allergen sensitization and challenge stage, modeling the natural route of allergen encounter (Fig. 3a). When we first tested LysM-Cre/Igf1fl/fl mice, these mice proved unsuitable, as we continued to detect variable levels of IGF-1 in the BAL fluid after HDM administration (likely due to leakage of serum IGF-1 during inflammation). Therefore, we targeted instead the IGF-1R on the epithelial cells. We crossed Igf1rfl/fl mice36 with CCSP-rtTA/tetO-Cre transgenic mice30,37, the latter driving Cre under the Club cell secretory protein (CCSP) promoter in the epithelial cells of the trachea, bronchi and bronchioles30,37. This CCSP-rtTA/tetO-Cre strain allows for inducible Cre expression through doxycycline administration via drinking water, thereby allowing normal development and gene deletion just prior to allergen exposure. We observed near complete loss of IGF-1R on epithelial cells of CCSP-Cre/Igf1rfl/fl mice after doxycycline treatment (Fig. 3b). After sensitization and challenge with low-endotoxin HDM, the CCSP-Cre/Igf1rfl/fl mice had greater airway inflammation based on several parameters. First, there was marked increase in eosinophils and CD4+ T-cells in the BAL fluid (Fig. 3c) and trending increase in inflammatory cells in lungs (Extended Data Fig. 6a). Second, lung-draining lymph nodes were larger with more CD4+ T-cells (Fig. 3d). Third, lung sections showed increased peribronchial and perivascular cellular infiltration (Fig. 3e, f) and greater mucus accumulation after HDM treatment (Fig. 3g, h). Fourth, HDM-treated CCSP-Cre/Igf1rfl/fl mice displayed increased airway reactivity after methacholine challenge, a measure of the bronchial hyper-responsiveness38,39 (Extended Data Fig. 6b). There were more apoptotic cells in lung sections (cleaved caspase 3 staining), likely due to greater inflammation (Extended Data Fig. 6c). These data suggested a requirement for IGF-1R in the airway epithelial cells in minimizing inflammation in this model.
These observations were initially surprising, as we were expecting the loss of IGF-1R on airway epithelial cells to improve apoptotic cell clearance and thereby attenuate, rather than worsen, inflammation. This prompted us to examine the temporal requirement of IGF-1/IGF-1R signaling in epithelial cells during allergen exposure. To distinguish the requirement for IGF-1/IGF-1R signaling at the sensitization versus challenge phase, we administered doxycycline at different times: deleting IGF-1R expression before allergen sensitization (Fig. 4a), or after the initial allergen sensitization but before the allergen challenge phase (Extended Data Fig. 7a). Mice with IGF-1R deletion prior to allergen sensitization showed a significant increase in inflammatory parameters (Fig. 4b), while deleting IGF-1R after the sensitization phase had minimal effect on disease severity (Extended Data Fig. 7b, c).
Airway epithelial cells encountering allergen can produce cytokines, such as TSLP, CSF-2/GM-CSF (affecting dendritic cell maturation), as well as IL-33 and IL-25 that drive type 2 innate lymphoid cells (ILC2s) to proliferate and produce IL-1340. Bronchial epithelial cells from human asthmatics produce elevated levels of IL-6, IL-8, and CSF-241, and TSLP released from airway epithelial cells (mice and human) can exacerbate airway inflammation42. When we assessed the cytokine profile in the BAL fluid of CCSP-Cre/Igf1rfl/fl mice sensitized with HDM, levels of IL-6 and TSLP were greater (Fig. 4d, e). We did not find increased levels of IL-33, as seen with Rac1 deletion in airway epithelial cells4, but this may be due to differences in inflammatory pathways regulated by the loss of IGF-1R versus Rac1. These results suggest that IGF-1R in airway epithelial cells limits inflammatory cytokines during the initial antigen exposure/sensitization phase.
As for trigger(s) for IGF-1 release from the alveolar macrophages in the sensitization phase, while IL-4 is often considered a classic Th2 cytokine from the adaptive immune response43,44, both IL-445–51 and IL-1340 can be produced during the allergen sensitization phase by resident mast cells, basophils, and/or ILC2s. In fact, IL-4 was enhanced (along with IL-5 and eotaxin-1) in the BAL fluid of CCSP-Cre/Igf1rfl/fl mice sensitized with HDM (Fig. 4c). Thus, IL-4 and/or apoptotic cells present during the initial allergen exposure could elicit IGF-1 production from alveolar macrophages.
Alveolar macrophages are reported to release microvesicles containing anti-inflammatory mediators during smoking-induced lung injury52. Since IGF-1 enhances liposome uptake by airway epithelial cells, we tested whether microvesicles from alveolar macrophages may impact airway epithelial cell response to HDM. Using differential centrifugation and filtration, we isolated and characterized microvesicles from alveolar macrophages (Fig. 4f). First, negative stain and cryo-electron microscopy revealed membrane bound spherical structures >100nm in diameter (Fig. 4g, h). Second, via ImagestreamX™ analysis, particles from an alveolar macrophage cell line or primary mouse alveolar macrophages carry markers of lung macrophage origin, CD11c or SiglecF, with variable Annexin V staining (Fig. 4i). Third, tunable resistive pulse sensing analysis of microvesicles revealed a mean particle size of 357±148.5nm (Fig. 4j), within the reported 100–1000nm range for microvesicles53. Further, IL-4 treatment of macrophages increased microvesicles secretion (Extended Data Fig. 8a). Also, uptake of these microvesicles by BEAS-2B airway epithelial cells was enhanced by IGF-1 (Fig. 4k). Importantly, adding alveolar macrophage derived microvesicles to HDM-treated BEAS-2B cells resulted in significantly lower TSLP, CSF2, IL6, and IL8 induction (Fig. 4l). To further explore this, we performed RNA-seq of BEAS-2B cells treated with HDM ± microvesicles. While HDM treatment increased several known genes associated with asthma in humans54–57, such as FGF2, KLF4, Interferon-Induced Protein with Tetratricopeptide Repeats 2 (IFIT2), and Pentraxin 3 (PTX3), adding microvesicles from alveolar macrophages suppressed transcription of these genes in the epithelial cells (Fig. 4m, Extended Data Fig. 8b). Thus, microvesicles released from alveolar macrophages acting on airway epithelial cells exposed to allergen could dampen airway inflammation.
The data presented here provide several new insights on phagocytosis and tissue inflammation. First, it has long been known that professional and non-professional phagocytes reside in proximity and can engulf dying cells and debris, yet communication between these phagocytes was not understood. The data presented here identifies a rapid, transient, and reversible regulation, wherein soluble IGF-1 from macrophages influences the type of particle uptake by epithelial cells. This transient effect might allow the macrophages to temporarily ‘redirect’ the non-professional phagocytes toward other function(s). Such a possible hierarchy in cell clearance could provide temporal and spatial cross-communication within a given tissue10. Second, our data identifies a two-part regulation of epithelial cells by macrophages which impacts airway inflammation, i.e., the secretion of IGF-1 that redirects particle uptake, and the release of microvesicles that dampen inflammatory cytokine production by epithelial cells (Extended Data Fig. 9). Third, IGF-1 is a growth factor widely linked to growth, cellular proliferation, and aging58–61. Global IGF-1 deletion in mice results in dwarfism and perinatal lethality, and IGF-1 mutations are linked to human diseases34,62,63. This work identifies a previously unappreciated biological function for the IGF-1/IGF-1R axis as a modulator of airway hyper-responsiveness to allergens with potential therapeutic relevance.
Methods
Mice
C57BL/6J, Igf1rfl/fl, and Igf1fl/fl mice were obtained from Jackson Laboratories. CCSP-rtTA/tetO-Cre were kindly provided by Dr. Jeffrey Whitsett at Cincinnati Children’s Hospital37. To generate IGF-1R deletion in Club cells, we crossed CCSP-rtTA/tetO-Cre mice to Igf1rfl/fl36. To achieve deletion, mice were given doxycycline (1mg/mL) in drinking water containing 0.4% sucrose for at least 7 days prior to beginning of allergen administration, unless otherwise noted. We also crossed Igf1fl/fl mice33,34 to LysM-Cre mice to conditionally delete Igf1 in the myeloid lineage. We have previously reported on the generation of CCSP-Cre/YFP mice4. For all in vivo experiments, except generation of apoptotic thymocytes, mice between the ages of 8 and 12 weeks were used. No blinding was performed for in vivo experiments. Mice were allocated to experimental groups based on genotype and age-matching. Female and male mice were used for all experiments, except for in vivo engulfment assays in which only male mice were used. Sample size was selected based on mice availability and power statistics. All animal procedures were performed according to the protocols provided by the Institutional Animal Care and Use Committee (IACUC) of the University of Virginia.
Induction of Airway Inflammation
Mice were given drinking water containing doxycycline (1mg/mL) seven days prior to first HDM administration. Mice were primed intranasally with 10μg of low endotoxin house dust mite extraction (Indoor Biotechnologies) on days 0, 2, 4 and then challenged intranasally on days 10, 12, and 14. On day 16, mice were harvested and analyzed for eosinophilic airway inflammation. Alternatively, mice were given three doses of low endotoxin HDM on days 0, 2, and 4 and analyzed on day 6 (“sensitization phase”). For the “challenge phase,” mice that had not received any doxycycline were given three doses of low endotoxin HDM on days 0, 2 and 4, then given doxycycline (via drinking water) from day 4 until the mice were analyzed. These mice were also challenged intranasally with three doses of low endotoxin HDM on days 10, 12, and 14 and analyzed on day 16. For airway inflammation experiments, sample size was selected to be above 3 mice per group per experiment.
Collection of BAL fluid, lymph nodes, and lung
For airway inflammation experiments, 0.8mL of PBS were delivered intratracheally through a cannula. Recovered BAL fluid was centrifuged and the supernatant was frozen at −80°C for subsequent Luminex analysis. Collected cells were stained for surface markers to distinguish macrophages, neutrophils, T-cells, and eosinophils. For lung harvests, mice were perfused through the right ventricle with PBS and the lungs were carefully excised and placed in type 2 collagenase (Worthington Biochemical Corporation) dissolved in HBSS containing Ca2+ and Mg2+. Lungs were minced and then incubated at 37°C for one hour, with vigorous pipetting to separate the tissue every 15 minutes. The lung homogenate was then passed through a 70μm nylon strainer, spun down and treated with red blood cell lysis buffer (Sigma-Aldrich) for 5 minutes. The cells were then washed and resuspended in PBS containing 0.1%BSA. Draining lymph nodes were carefully extracted, and a single cell suspension was made by passage through a 70μm nylon strainer using the flat end of a syringe. Cells were washed and then resuspended in PBS containing 0.1%BSA.
Cell staining and total cell numbers
The collected cells were stained for macrophages, neutrophils, T cells, and eosinophils using the following markers: CD11c (eBioscience, cl. N418), Siglec F (BD Biosciences, cl. E50-2440), Ly6G (eBioscience, clone 1A8), CD11b (eBioscience, cl. M1/70), F4/80 (eBioscience, cl. BM8), CD3 (eBioscience, cl. 145-2C11), CD4 (eBioscience, cl. RM4-5), CD44 (eBioscience, cl. IM7), CD69 (eBioscience, cl. H1.2F3). Absolute cell numbers were determined using AccuCount Particles (Spherotech). Flow cytometry data was collected on FACS Canto I (Becton Dickinson) and analyzed with FlowJo (Treestar, Inc).
Microscopy and histology
For hematoxylin and eosin and Periodic acid Schiff staining of lung sections, mice were perfused with PBS and a cannula inserted into the trachea. The lungs were gently inflated with 10% formalin at a constant fluid pressure at 25 cm. The trachea was tied off and the entire heart and lung were removed and placed in 10% formalin. Lungs were paraffin embedded, sectioned and stained by HistoTox Labs (Boulder, CO). Additional lung sections were embedded and sectioned by Research Histology Core at University of Virginia and the immunohistochemical staining for IGF-1R, and cleaved caspase 3 was performed by the University of Virginia Biorepository and Tissue Research Facility. Approximately 6–10 images were taken per mice, with a total of 3–4 mice per group, and blindly scored by two independent scorers for inflammation, PAS staining, and cleaved caspase 3 positive cells. Immunofluorescence staining of lung sections was performed at the University of Virginia Cardiovascular Research Center Histological Services.
Airway hypersensitivity
Mice were anesthetized and given a tracheotomy tube that delivered increasing concentrations of aerosolized methacholine. The tracheotomy tube in turn was connected to the inspiratory and expiratory ports of a volume-cycled ventilator (flexiVent; SCIREQ Scientific). Airway resistance was measured at baseline and after each dose of methacholine.
Macrophage isolation
To obtain bone-marrow derived macrophages, femurs were removed from 8 week old mice and flushed with 5mL of sterile PBS containing 5% FBS. The cell suspension was centrifuged, treated with red blood cell lysis buffer, washed, and then plated onto sterile petri dishes in DMEM containing 10% L929 media, 10% FBS and 1% penicillin/streptomycin/glutamine (PSQ). Media was replenished every 2 to 3 days and differentiated cells were used at day 6 post-harvest. Resident peritoneal macrophages were obtained by flushing the peritoneal cavity of mice with 10mL of cold PBS containing 5% FBS. Collected cells were spun down, resuspended in X-VIVO 10 (Lonza) and plated at a concentration of 3×105 cells per well in a 24 well plate for IGF-1 secretion assays, and 5×105 per well in a 24 well plate for engulfment assays. Floating cells were washed the next day and remaining peritoneal macrophages were used 2 days after isolation. Alveolar macrophages were isolated by flushing the lungs with 1mL of cold PBS instilled intratracheally (five flushes). Collected cells were centrifuged, resuspended in F12K media containing 10% FBS and 1% PSQ, and seeded at 1×105 cells per well in a 48-well plate. All floating cells were washed away the next day and remaining cells were used in assays 2 days after isolation.
Microvesicle isolation
MH-S, mouse alveolar macrophages, or primary mouse alveolar macrophages were seeded. After adherence, the media was replaced with .22μm filtered media to remove any contaminating microvesicles. After an overnight incubation, the supernatant was harvested and spun at 5000xg to remove cell debris and apoptotic bodies. The pellet was discarded and the resulting supernatant was filtered through 0.8μm filter and spun again at 17,000xg. The pelleted microvesicles were then washed with HBSS and then spun again at 17,000xg. For engulfment assays, microvesicles were stained with TAMRA for 20 minutes and then added to BEAS-2B cells for 90 minutes. For flow cytometry, purified particles were stained with CD11c, Siglec F, and Annexin V (BD Biosciences, Cat. No. 550475) and processed on ImagestreamX™ imaging flow cytometer (Amnis). Microvesicle size distribution was characterized using qNano (IZON Science) with a NP400 membrane and at least 500 particles were counted. Microvesicles were prepared for cryo-electron microscopy using standard methods and imaged on an FEI TF2064.
In vivo cytokine or apoptotic cell administration
Mice were administered intranasally with 1μg of recombinant mouse IL-4, IL-5, or IL-13 (eBiosciences), 1×106 apoptotic Jurkat cells, or PBS as control, for two consecutive days. On the third day, BAL fluid was recovered and centrifuged; the supernatant was stored at −80°C for subsequent cytokine analysis. For cytokine or apoptotic cell administration, sample size was selected to be 2 or more mice per group per experiment.
Cytokine and IGF-1 analysis
IL-4, IL-5, IL-6, and CCL-11 in the BAL fluid of mice that only received priming with HDM (regimen #1) were quantified by a multiplex Luminex performed by the University of Virginia Flow Cytometry Core Facility. Secretion of IGF-1 from J774 cells, peritoneal macrophages, and BAL fluids, as well as TSLP from BAL fluids, were measured by ELISA (R&D Systems).
In vitro cell systems
LR73 (hamster fibroblasts), SVEC-40 (mouse endothelial cells), BEAS-2B (human bronchial epithelial cells, ATCC #CRL-9609), 16HBE14o- cells (human bronchial epithelial cells), MH-S (mouse alveolar macrophage cells ATCC #CRL-2019), Jurkat (human T-cells) were either available in the laboratory or obtained from ATCC, with the latter tested for mycoplasma.
In vitro engulfment assay
LR73, SVEC-40, BEAS-2B, and 16HBE14o- cells were seeded in a 24 well plate. Thymocytes were isolated from 4–6 week old mice and induced to undergo apoptosis with dexamethasone. Thymocytes or prepared microvesicles were then stained with either CypHer5E (GE Healthcare, PA15401) or TAMRA (Invitrogen, C-1171). LR73 and SVEC-40 cells were incubated with apoptotic thymocytes at a 1:10 phagocyte to target ratio, BEAS-2B at a 1:5 phagocyte to target ratio, and 16HBE14o- at a 1:20 phagocyte to target ratio for 2 hours. Mouse IGF-1 (Sigma-Aldrich), human IGF-1 (Sigma-Aldrich), human IGF-II (Sigma-Aldrich) were added to the phagocytes at the same time as addition of apoptotic targets. For IGFBP3 studies, IGFBP3 (Sigma-Aldrich) was added to media or supernatant from J774 cells for one hour to allow IGFBP3 to bind to any available IGF-1, then the mix is added to phagocytes along with apoptotic targets. For all pharmacological studies, phagocytes were pre-incubated with the compounds listed below for one hour prior to addition of apoptotic targets: cytochalasin D (Sigma-Aldrich, C8273, 1μM), Latrunculin A (Tocris, 3973, 150nM), CK-666 (Tocris, 3950, 25μM-100μM), OSI-906 (Selleckchem, S1091, 5nM-40nM), NVP-AEW541 (Selleckchem, S1034, 25nM-100nM), Rapamycin (Sigma-Aldrich, R0395, 10μM-1mM), MK-2206 2HCl (Selleckchem, S1078, 10nM-1μM), U0126-EtOH (Selleckchem, S1102, 8pM-5nM), Wortmannin (Sigma-Aldrich, W1628, 50nM-200nM), Y27632 (Calbiochem, 688000, 3.75μM-15μM), or GSK269962 (Tocris, 4009, 80nM-2μM). Phagocytes were examined to ensure no gross morphological changes occurred due to drug treatment. Targets were then washed off three times with PBS, and the cells were dissociated from the plate with trypsin and the engulfment assessed by flow cytometry.
In vivo engulfment assay
CCSP-Cre/YFP mice were administered PBS or 1μg of IGF-1 intranasally. One hour later, 100 million CypHer5E-labelled apoptotic thymocytes with or without IGF-1 were injected intranasally for 3.5 hours. The BAL fluid was harvested and the lungs excised, minced, and digested into a single cell suspension. The cells were then stained with appropriate markers: CD11c and Siglec F for alveolar macrophage markers and EpCam (along with YFP expression) for airway epithelial cells and analyzed by flow cytometry to assess apoptotic cell uptake by the airway epithelial cells and the alveolar macrophages.
Liposome construction
Liposomes were prepared by dissolving the lipids (phosphatidylserine, dioleoyl phosphatidylcholine, cholesterol and the lipid DiD dye) in chloroform, evaporating chloroform under flow of argon gas in a glass vial, then subjecting the lipid layer to overnight lyophilization to remove traces of organic solvent. Then normal saline was added for hydration, and intense vortexing was preformed to prepare multilamellar vesicles (MLV). Liposomes were repeatedly filtered through a 0.2um Nuclepore polycarbonate filter to prepare smaller particles. Particle size was verified by dynamic light scattering using Nicomp 370.
Immunoblotting
LR73, J774 or BEAS-2B cells were seeded in a 60mm dish at a concentration of 5×105. Cells were serum-starved for 6 hours and then stimulated with 100ng/mL of IGF-1 for various time points. Cells were lysed in RIPA buffer and used in Western blots. The blots were probed for phospho-Erk1/2 (Cell Signaling Technology, #4370), phospho-Akt (Cell Signaling Technology, #4060), phospho-IGF-1R (Cell Signaling, #3024), total Erk2 (Santa Cruz Biotechnology, #sc-154-G), total Akt (Cell Signaling Technology, #4691), total IGF-1R (Cell Signaling, #9750), and anti-B-actin-HRP (Sigma-Aldrich, #A3854) followed by chemiluminescence detection.
Quantitative RT-PCR
Total RNA was extracted from cells using Quick-RNA Miniprep Kit (Zymo Research) or RNeasy Mini Kit (Qiagen) and cDNA was synthesized using QuantiTect Reverse Transcription Kit (Qiagen) according to manufacturers’ instructions. Quantitative gene expression for mouse Igf1, human TSLP, CSF2, IL6, IL8 or housekeeping human or mouse Hprt was performed using Taqman probes (Applied Biosystems) using StepOnePlus Real Time PCR System (ABI).
RNA-seq
BEAS-2B cells were treated with HDM and alveolar macrophage-derived microvesicles for 3 hours. Total RNA was extracted and an mRNA library was prepared using Illumina TruSeq platform and followed by transcriptome sequencing using an Illumina NextSeq 500 cartridge. Rv3.2.2 was used for graphical and statistical analysis and the R package DESeq2 was used for differential gene expression analysis of RNA-seq data. RNA-seq analysis was performed by the UVa Bioinformatics Core.
Code Availability
R code used for bioinformatics analysis and heat map generation is available upon request.
Statistical analysis
Statistical significance was determined using GraphPad Prism 5 or 6 using unpaired Student’s two-tailed t-test, one-sample t-test, one-way ANOVA or two-way ANOVA, as according to test requirements. Grubbs’ Outlier Test was used to determine outliers, which were excluded from final analysis. A p-value of <0.05 (indicated by one asterisk), <0.01 (indicated by two asterisks), or <0.001 (indicated by three asterisks) were considered significant.
Extended Data
Acknowledgments
The authors thank members of the Ravichandran laboratory for discussions and critical reading of manuscript; Sanja Arandjelovic, Kristen Penberthy, and Lisa Haney for scoring histology slides, and Justin Perry for assistance with generation of the heat map, and the UVa flow cytometry core and the UVa Molecular electron microscopy core. This work is supported by grants to K.S.R. from the NIGMS (GM064709), NIMH (MH096484), NHLBI (P01HL120840), and Center for Cell Signaling at the University of Virginia, NHLBI (HL132287 and HL091127) to Y.M.S. Additional support was provided via the NIH Training Grants T32 GM008136 (Cell and Molecular Biology) to C.Z.H.; T32 AI007496 (Immunology) to C.Z.H., M.W.B., and I.J.J.; and T32 GM007267 (MSTP) to M.W.B.
Footnotes
Author Contributions: authors are required to include a statement to specify the contributions of each co-author. The statement can be up to several sentences long, describing the tasks of individual authors referred to by their initials. See the authorship policy page for further explanation and examples.
C.Z.H. designed, performed and analyzed most of the experiments in this study with input from K.S.R. I.J.J., J.M.K., M.W.B. assisted with in vivo experiments. A.L.K. provided phosphatidylserine liposomes. S.O-G ran the RNA-seq. U.E. performed microvesicle quantification using qNano. Y.M.S. performed airway resistance experiments. K.S.T. assisted in evaluation of lung pathology. C.Z.H. and K.S.R. wrote the manuscript with input from co-authors.
The authors declare no competing financial interests.
References
- 1.Nagata S, Hanayama R, Kawane K. Autoimmunity and the clearance of dead cells. Cell. 2010;140:619–630. doi: 10.1016/j.cell.2010.02.014. [DOI] [PubMed] [Google Scholar]
- 2.Henson PM, Hume DA. Apoptotic cell removal in development and tissue homeostasis. Trends Immunol. 2006;27:244–250. doi: 10.1016/j.it.2006.03.005. [DOI] [PubMed] [Google Scholar]
- 3.Burstyn-Cohen T, et al. Genetic dissection of TAM receptor-ligand interaction in retinal pigment epithelial cell phagocytosis. Neuron. 2012;76:1123–1132. doi: 10.1016/j.neuron.2012.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Juncadella IJ, et al. Apoptotic cell clearance by bronchial epithelial cells critically influences airway inflammation. Nature. 2013;493:547–551. doi: 10.1038/nature11714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Wood W, et al. Mesenchymal cells engulf and clear apoptotic footplate cells in macrophageless PU.1 null mouse embryos. Development. 2000;127:5245–5252. doi: 10.1242/dev.127.24.5245. [DOI] [PubMed] [Google Scholar]
- 6.Lu Z, et al. Phagocytic activity of neuronal progenitors regulates adult neurogenesis. Nat Cell Biol. 2011;13:1076–1083. doi: 10.1038/ncb2299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ichimura T, et al. Kidney injury molecule-1 is a phosphatidylserine receptor that confers a phagocytic phenotype on epithelial cells. J Clin Invest. 2008;118:1657–1668. doi: 10.1172/JCI34487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Mesa KR, et al. Niche-induced cell death and epithelial phagocytosis regulate hair follicle stem cell pool. Nature. 2015;522:94–97. doi: 10.1038/nature14306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Elliott MR, et al. Unexpected requirement for ELMO1 in clearance of apoptotic germ cells in vivo. Nature. 2010;467:333–337. doi: 10.1038/nature09356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Gregory CD, Pound JD. Microenvironmental influences of apoptosis in vivo and in vitro. Apoptosis. 2010;15:1029–1049. doi: 10.1007/s10495-010-0485-9. [DOI] [PubMed] [Google Scholar]
- 11.Arandjelovic S, Ravichandran KS. Phagocytosis of apoptotic cells in homeostasis. Nat Immunol. 2015;16:907–917. doi: 10.1038/ni.3253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Green DR. The end and after: how dying cells impact the living organism. Immunity. 2011;35:441–444. doi: 10.1016/j.immuni.2011.10.003. [DOI] [PubMed] [Google Scholar]
- 13.Werner S, Grose R. Regulation of wound healing by growth factors and cytokines. Physiol Rev. 2003;83:835–870. doi: 10.1152/physrev.00031.2002. [DOI] [PubMed] [Google Scholar]
- 14.Yakar S, et al. Circulating levels of IGF-1 directly regulate bone growth and density. J Clin Invest. 2002;110:771–781. doi: 10.1172/JCI15463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Firth SM, Baxter RC. Cellular actions of the insulin-like growth factor binding proteins. Endocr Rev. 2002;23:824–854. doi: 10.1210/er.2001-0033. [DOI] [PubMed] [Google Scholar]
- 16.Mulvihill MJ, et al. Discovery of OSI-906: a selective and orally efficacious dual inhibitor of the IGF-1 receptor and insulin receptor. Future Med Chem. 2009;1:1153–1171. doi: 10.4155/fmc.09.89. [DOI] [PubMed] [Google Scholar]
- 17.Garcia-Echeverria C, et al. In vivo antitumor activity of NVP-AEW541-A novel, potent, and selective inhibitor of the IGF-IR kinase. Cancer Cell. 2004;5:231–239. doi: 10.1016/s1535-6108(04)00051-0. [DOI] [PubMed] [Google Scholar]
- 18.Fernandez AM, Torres-Aleman I. The many faces of insulin-like peptide signalling in the brain. Nat Rev Neurosci. 2012;13:225–239. doi: 10.1038/nrn3209. [DOI] [PubMed] [Google Scholar]
- 19.LeRoith D. Insulin-like growth factor I receptor signaling--overlapping or redundant pathways? Endocrinology. 2000;141:1287–1288. doi: 10.1210/endo.141.4.7475. [DOI] [PubMed] [Google Scholar]
- 20.Tosello-Trampont AC, Nakada-Tsukui K, Ravichandran KS. Engulfment of apoptotic cells is negatively regulated by Rho-mediated signaling. J Biol Chem. 2003;278:49911–49919. doi: 10.1074/jbc.M306079200. [DOI] [PubMed] [Google Scholar]
- 21.Leverrier Y, Ridley AJ. Requirement for Rho GTPases and PI 3-kinases during apoptotic cell phagocytosis by macrophages. Curr Biol. 2001;11:195–199. doi: 10.1016/s0960-9822(01)00047-1. [DOI] [PubMed] [Google Scholar]
- 22.Tosello-Trampont AC, Brugnera E, Ravichandran KS. Evidence for a conserved role for CRKII and Rac in engulfment of apoptotic cells. J Biol Chem. 2001;276:13797–13802. doi: 10.1074/jbc.M011238200. [DOI] [PubMed] [Google Scholar]
- 23.Schlam D, et al. Phosphoinositide 3-kinase enables phagocytosis of large particles by terminating actin assembly through Rac/Cdc42 GTPase-activating proteins. Nat Commun. 2015;6:8623. doi: 10.1038/ncomms9623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Casella JF, Flanagan MD, Lin S. Cytochalasin D inhibits actin polymerization and induces depolymerization of actin filaments formed during platelet shape change. Nature. 1981;293:302–305. doi: 10.1038/293302a0. [DOI] [PubMed] [Google Scholar]
- 25.Yarmola EG, Somasundaram T, Boring TA, Spector I, Bubb MR. Actin-latrunculin A structure and function. Differential modulation of actin-binding protein function by latrunculin A. J Biol Chem. 2000;275:28120–28127. doi: 10.1074/jbc.M004253200. [DOI] [PubMed] [Google Scholar]
- 26.Hetrick B, Han MS, Helgeson LA, Nolen BJ. Small molecules CK-666 and CK-869 inhibit actin-related protein 2/3 complex by blocking an activating conformational change. Chem Biol. 2013;20:701–712. doi: 10.1016/j.chembiol.2013.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chen F, et al. An essential role for TH2-type responses in limiting acute tissue damage during experimental helminth infection. Nat Med. 2012;18:260–266. doi: 10.1038/nm.2628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Wynes MW, Frankel SK, Riches DW. IL-4-induced macrophage-derived IGF-I protects myofibroblasts from apoptosis following growth factor withdrawal. J Leukoc Biol. 2004;76:1019–1027. doi: 10.1189/jlb.0504288. [DOI] [PubMed] [Google Scholar]
- 29.Ginhoux F, Guilliams M. Tissue-Resident Macrophage Ontogeny and Homeostasis. Immunity. 2016;44:439–449. doi: 10.1016/j.immuni.2016.02.024. [DOI] [PubMed] [Google Scholar]
- 30.Perl AK, Tichelaar JW, Whitsett JA. Conditional gene expression in the respiratory epithelium of the mouse. Transgenic Res. 2002;11:21–29. doi: 10.1023/a:1013986627504. [DOI] [PubMed] [Google Scholar]
- 31.Wills-Karp M. Interleukin-13 in asthma pathogenesis. Curr Allergy Asthma Rep. 2004;4:123–131. doi: 10.1007/s11882-004-0057-6. [DOI] [PubMed] [Google Scholar]
- 32.Hilton DJ, et al. Cloning and characterization of a binding subunit of the interleukin 13 receptor that is also a component of the interleukin 4 receptor. Proc Natl Acad Sci U S A. 1996;93:497–501. doi: 10.1073/pnas.93.1.497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Liu JL, Yakar S, LeRoith D. Conditional knockout of mouse insulin-like growth factor-1 gene using the Cre/loxP system. Proc Soc Exp Biol Med. 2000;223:344–351. doi: 10.1046/j.1525-1373.2000.22349.x. [DOI] [PubMed] [Google Scholar]
- 34.Liu JL, et al. Insulin-like growth factor-I affects perinatal lethality and postnatal development in a gene dosage-dependent manner: manipulation using the Cre/loxP system in transgenic mice. Mol Endocrinol. 1998;12:1452–1462. doi: 10.1210/mend.12.9.0162. [DOI] [PubMed] [Google Scholar]
- 35.Thomas WR, Hales BJ, Smith WA. House dust mite allergens in asthma and allergy. Trends Mol Med. 2010;16:321–328. doi: 10.1016/j.molmed.2010.04.008. [DOI] [PubMed] [Google Scholar]
- 36.Dietrich P, Dragatsis I, Xuan S, Zeitlin S, Efstratiadis A. Conditional mutagenesis in mice with heat shock promoter-driven cre transgenes. Mamm Genome. 2000;11:196–205. doi: 10.1007/s003350010037. [DOI] [PubMed] [Google Scholar]
- 37.Perl AK, Zhang L, Whitsett JA. Conditional expression of genes in the respiratory epithelium in transgenic mice: cautionary notes and toward building a better mouse trap. Am J Respir Cell Mol Biol. 2009;40:1–3. doi: 10.1165/rcmb.2008-0011ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hahn YS, et al. V gamma 4+ gamma delta T cells regulate airway hyperreactivity to methacholine in ovalbumin-sensitized and challenged mice. J Immunol. 2003;171:3170–3178. doi: 10.4049/jimmunol.171.6.3170. [DOI] [PubMed] [Google Scholar]
- 39.Townley RG, Horiba M. Airway hyperresponsiveness: a story of mice and men and cytokines. Clin Rev Allergy Immunol. 2003;24:85–110. doi: 10.1385/CRIAI:24:1:85. [DOI] [PubMed] [Google Scholar]
- 40.Lambrecht BN, Hammad H. The immunology of asthma. Nat Immunol. 2015;16:45–56. doi: 10.1038/ni.3049. [DOI] [PubMed] [Google Scholar]
- 41.Marini M, Vittori E, Hollemborg J, Mattoli S. Expression of the potent inflammatory cytokines, granulocyte-macrophage-colony-stimulating factor and interleukin-6 and interleukin-8, in bronchial epithelial cells of patients with asthma. J Allergy Clin Immunol. 1992;89:1001–1009. doi: 10.1016/0091-6749(92)90223-o. [DOI] [PubMed] [Google Scholar]
- 42.Zhou B, et al. Thymic stromal lymphopoietin as a key initiator of allergic airway inflammation in mice. Nat Immunol. 2005;6:1047–1053. doi: 10.1038/ni1247. [DOI] [PubMed] [Google Scholar]
- 43.Lambrecht BN, Hammad H. The immunology of asthma. Nat Immunol. 2014;16:45–56. doi: 10.1038/ni.3049. [DOI] [PubMed] [Google Scholar]
- 44.Barnes PJ. Immunology of asthma and chronic obstructive pulmonary disease. Nat Rev Immunol. 2008;8:183–192. doi: 10.1038/nri2254. [DOI] [PubMed] [Google Scholar]
- 45.Machado DC, Horton D, Harrop R, Peachell PT, Helm BA. Potential allergens stimulate the release of mediators of the allergic response from cells of mast cell lineage in the absence of sensitization with antigen-specific IgE. Eur J Immunol. 1996;26:2972–2980. doi: 10.1002/eji.1830261224. [DOI] [PubMed] [Google Scholar]
- 46.Huels C, et al. Co-activation of naive CD4+ T cells and bone marrow-derived mast cells results in the development of Th2 cells. Int Immunol. 1995;7:525–532. doi: 10.1093/intimm/7.4.525. [DOI] [PubMed] [Google Scholar]
- 47.Bradding P, et al. TNF alpha is localized to nasal mucosal mast cells and is released in acute allergic rhinitis. Clin Exp Allergy. 1995;25:406–415. doi: 10.1111/j.1365-2222.1995.tb01071.x. [DOI] [PubMed] [Google Scholar]
- 48.Gessner A, Mohrs K, Mohrs M. Mast cells, basophils, and eosinophils acquire constitutive IL-4 and IL-13 transcripts during lineage differentiation that are sufficient for rapid cytokine production. J Immunol. 2005;174:1063–1072. doi: 10.4049/jimmunol.174.2.1063. [DOI] [PubMed] [Google Scholar]
- 49.Sokol CL, Barton GM, Farr AG, Medzhitov R. A mechanism for the initiation of allergen-induced T helper type 2 responses. Nat Immunol. 2008;9:310–318. doi: 10.1038/ni1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sokol CL, Medzhitov R. Role of basophils in the initiation of Th2 responses. Curr Opin Immunol. 2010;22:73–77. doi: 10.1016/j.coi.2010.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sokol CL, Medzhitov R. Emerging functions of basophils in protective and allergic immune responses. Mucosal Immunol. 2010;3:129–137. doi: 10.1038/mi.2009.137. [DOI] [PubMed] [Google Scholar]
- 52.Bourdonnay E, et al. Transcellular delivery of vesicular SOCS proteins from macrophages to epithelial cells blunts inflammatory signaling. J Exp Med. 2015;212:729–742. doi: 10.1084/jem.20141675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Burger D, et al. Microparticles: biomarkers and beyond. Clin Sci (Lond) 2013;124:423–441. doi: 10.1042/CS20120309. [DOI] [PubMed] [Google Scholar]
- 54.Bissonnette EY, et al. Fibroblast growth factor-2 is a sputum remodeling biomarker of severe asthma. J Asthma. 2014;51:119–126. doi: 10.3109/02770903.2013.860164. [DOI] [PubMed] [Google Scholar]
- 55.Zhang J, et al. Pentraxin 3 (PTX3) expression in allergic asthmatic airways: role in airway smooth muscle migration and chemokine production. PLoS One. 2012;7:e34965. doi: 10.1371/journal.pone.0034965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Yang IV, et al. The clinical and environmental determinants of airway transcriptional profiles in allergic asthma. Am J Respir Crit Care Med. 2012;185:620–627. doi: 10.1164/rccm.201108-1503OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Chamberland A, Madore AM, Tremblay K, Laviolette M, Laprise C. A comparison of two sets of microarray experiments to define allergic asthma expression pattern. Exp Lung Res. 2009;35:399–410. doi: 10.1080/01902140902745174. [DOI] [PubMed] [Google Scholar]
- 58.Yakar S, Liu JL, Le Roith D. The growth hormone/insulin-like growth factor-I system: implications for organ growth and development. Pediatr Nephrol. 2000;14:544–549. doi: 10.1007/s004670000363. [DOI] [PubMed] [Google Scholar]
- 59.Musaro A, et al. Localized Igf-1 transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle. Nat Genet. 2001;27:195–200. doi: 10.1038/84839. [DOI] [PubMed] [Google Scholar]
- 60.Kenyon CJ. The genetics of ageing. Nature. 2010;464:504–512. doi: 10.1038/nature08980. [DOI] [PubMed] [Google Scholar]
- 61.Lin K, Hsin H, Libina N, Kenyon C. Regulation of the Caenorhabditis elegans longevity protein DAF-16 by insulin/IGF-1 and germline signaling. Nat Genet. 2001;28:139–145. doi: 10.1038/88850. [DOI] [PubMed] [Google Scholar]
- 62.Gasparini L, Xu H. Potential roles of insulin and IGF-1 in Alzheimer’s disease. Trends Neurosci. 2003;26:404–406. doi: 10.1016/S0166-2236(03)00163-2. [DOI] [PubMed] [Google Scholar]
- 63.Smith TJ. Insulin-like growth factor-I regulation of immune function: a potential therapeutic target in autoimmune diseases? Pharmacol Rev. 2010;62:199–236. doi: 10.1124/pr.109.002469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Yeager M, Berriman JA, Baker TS, Bellamy AR. Three-dimensional structure of the rotavirus haemagglutinin VP4 by cryo-electron microscopy and difference map analysis. EMBO J. 1994;13:1011–1018. doi: 10.1002/j.1460-2075.1994.tb06349.x. [DOI] [PMC free article] [PubMed] [Google Scholar]