[go: up one dir, main page]
More Web Proxy on the site http://driver.im/ Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2017 Mar 14;114(13):E2758–E2765. doi: 10.1073/pnas.1611665114

Receptor Mincle promotes skin allergies and is capable of recognizing cholesterol sulfate

Alexey V Kostarnoy a,1, Petya G Gancheva a, Bernd Lepenies b, Amir I Tukhvatulin c, Alina S Dzharullaeva c, Nikita B Polyakov d,e, Daniil A Grumov d, Daria A Egorova a, Andrey Y Kulibin f, Maxim A Bobrov g, Ekaterina A Malolina f,h, Pavel A Zykin i, Andrey I Soloviev d, Evgeniy Riabenko j,k, Diana V Maltseva l, Dmitry A Sakharov l, Alexander G Tonevitsky m, Lyudmila V Verkhovskaya n, Denis Y Logunov c, Boris S Naroditsky a, Alexander L Gintsburg o
PMCID: PMC5380039  PMID: 28292894

Significance

Post-traumatic sterile inflammation is the first necessary step of wound healing. In addition, sterile inflammation underlies the pathogenesis of a multitude of common diseases, such as allergies and autoimmune diseases. The molecular mechanisms underlying sterile inflammation are still not fully understood. Here, we show that the receptor Mincle (Clec4e), the expression of which is highly induced in the skin in response to damage, recognizes cholesterol sulfate, a molecule that is abundant in the epidermal layer of the skin, subsequently inducing a pro-inflammatory response. We also identify a role for Mincle as a driving component in the pathogenesis of allergic skin inflammation. The results demonstrate a previously unconsidered important role of Mincle in mediating sterile inflammation.

Keywords: innate immunity, sterile inflammation, allergy, Mincle, cholesterol sulfate

Abstract

Sterile (noninfected) inflammation underlies the pathogenesis of many widespread diseases, such as allergies and autoimmune diseases. The evolutionarily conserved innate immune system is considered to play a key role in tissue injury recognition and the subsequent development of sterile inflammation; however, the underlying molecular mechanisms are not yet completely understood. Here, we show that cholesterol sulfate, a molecule present in relatively high concentrations in the epithelial layer of barrier tissues, is selectively recognized by Mincle (Clec4e), a C-type lectin receptor of the innate immune system that is strongly up-regulated in response to skin damage. Mincle activation by cholesterol sulfate causes the secretion of a range of proinflammatory mediators, and s.c. injection of cholesterol sulfate results in a Mincle-mediated induction of a severe local inflammatory response. In addition, our study reveals a role of Mincle as a driving component in the pathogenesis of allergic skin inflammation. In a well-established model of allergic contact dermatitis, the absence of Mincle leads to a significant suppression of the magnitude of the skin inflammatory response as assessed by changes in ear thickness, myeloid cell infiltration, and cytokine and chemokine secretion. Taken together, our results provide a deeper understanding of the fundamental mechanisms underlying sterile inflammation.


The recognition of tissue injury and the subsequent responses leading to (i) the neutralization and elimination of the agent causing the damage and (ii) the repair of damaged tissues are essential challenges faced by multicellular organisms. In higher organisms, injuries and infections induce inflammatory responses. Inflammation plays roles in the localization and elimination of injurious agents, the removal of damaged tissues, and the initiation of the repair process. Evolutionarily conserved pattern recognition receptors (PRRs) of the innate immune system are capable of recognizing molecules associated with pathogens (pathogen-associated molecular patterns, or PAMPs) and endogenous molecules released from stressed or damaged cells in response to injury (damage-associated molecular patterns, DAMPs) (1). Recognizing the ligands of PRRs of either exogenous or endogenous origin subsequently induces an inflammatory response. Numerous DAMPs have been proposed (1, 2). However, the ability of many of these compounds to induce sterile inflammation is still under debate because some of them display activities at nonphysiological concentrations, and others are present in high abundance as components of the extracellular matrix (3, 4).

Posttraumatic inflammation is the first necessary step of wound healing (5). Nevertheless, a prolonged sterile (noninfected) inflammatory response underlies the pathogenesis of a multitude of widespread diseases, such as allergic diseases, autoimmune diseases, atherosclerosis, and cancer. Although progress has been made in identifying potential DAMPs, the molecular mechanisms underlying the induction of sterile inflammation are still not fully understood. This knowledge gap is well illustrated by the fact that for a number of the widespread diseases associated with acute or chronic sterile inflammation, endogenous molecular inductors of the inflammatory process and their receptors remain to be fully elucidated, and there are still either no or very limited effective therapies available for these diseases.

In the present study, we initially investigated changes in the gene expression profile of PRRs after skin injury in a mouse model. We determined that, among the different PRRs, the receptor Mincle was the most inducible in response to skin trauma. Mincle (also called Clec4e or Clecsf9) is a type-II transmembrane C-type lectin receptor that recognizes mycobacteria and pathogenic fungi. PAMPs recognized by Mincle are predominantly amphiphilic lipids, such as trehalose-6,6′-dimycolate (TDM) from Mycobacterium tuberculosis and glyceroglycolipids from Malassezia fungi (6). Mincle also recognizes endogenous spliceosome-associated protein 130 (SAP130), which is a constitutively expressed nuclear protein and a subunit of the histone deacetylase complex (7). The ligation of Mincle by its ligands leads to the interaction of Mincle with the adaptor protein Fc receptor γ-chain (FcRγ) and the recruitment of spleen tyrosine kinase (Syk) (7, 8). The signal is subsequently transmitted to NF-κB via the CARD9-BCL10-MALT1 pathway, leading to the production of a spectrum of proinflammatory cytokines (6, 8, 9).

We determined that after skin wounding or after a single topical application of the irritant and sensitizer 2,4-dinitrofluorobenzene (DNFB), the early secretion of proinflammatory mediators in the absence of Mincle was significantly impaired. Assuming that Mincle may be an important sensor of tissue damage, we performed a search for new endogenous ligands of this receptor. Using chromatography and mass spectrometry, we found that cholesterol sulfate, an abundant molecule in the epithelial barrier layers in the skin and gastrointestinal and respiratory tracts (1012), is selectively recognized by Mincle. Here, we show that Mincle activation by cholesterol sulfate causes the secretion of a range of proinflammatory mediators and that s.c. injection of cholesterol sulfate results in a Mincle-mediated induction of a severe local inflammatory response.

In the study, we identified a role of Mincle in the development of the pathologic process of cutaneous allergic inflammation. In a mouse model of allergic contact dermatitis, contact hypersensitivity (CHS) was induced via the topical application of the DNFB. Using Mincle receptor-knockout (Mincle-KO) mice, we observed that the absence of Mincle leads to a strong suppression of skin inflammation as reflected by changes in swelling, myeloid cell infiltration, and cytokine and chemokine secretion.

Collectively, these findings demonstrate an important role of the receptor Mincle in mediating sterile (noninfected) skin inflammation and provide a deeper understanding of the fundamental mechanisms underlying sterile inflammation.

Results

Mincle Is a PRR in the Skin That Is Highly Inducible in Response to Trauma and Mediates the Early Inflammatory Response.

We first assessed changes in the gene expression profiles of the receptors involved in innate responses to skin injuries in mice. We modeled excisional cutaneous wounds using a punch biopsy instrument. Wounds were made through the epidermis, dermis, and s.c. tissue layers, leaving the fascia intact. Skin tissue samples containing the wounds were removed 24 h and 4 d after injury, and intact skin was used as a control. RNA was extracted from samples, and a whole-genome analysis was performed using an Affymetrix microarray platform (Agilent Technologies). The results revealed that skin injury induced an increase in the expression of a number of PRR genes belonging to different families of innate immunity receptors, such as Toll-like receptors (TLRs), C-type lectin receptors (CLRs), NOD-like receptors (NLRs), and RIG-I–like receptors (RLRs). The fold-changes in the transcript levels of innate immune PRR genes in wounded skin relative to levels in intact skin are presented in Fig. 1A (the complete dataset has been deposited in the GEO database under accession no. GSE76144). Interestingly, one of the genes of the CLR family, Clec4e (Mincle), was extremely inducible after wounding in comparison with the changes in the other innate immune receptors. Another gene of the C-type lectin receptor family, Clec4d (MCL), which is located in the CLR cluster next to Clec4e and is considered to have emerged via a gene duplication of Clec4e (13), was the second most inducible gene at both time points among the innate immune system receptor genes.

Fig. 1.

Fig. 1.

Mincle (Clec4e) is a PRR in the skin that is highly inducible in response to trauma and mediates the early inflammatory response. (A) Whole-genome microarray analysis of the changes in gene expression of innate immune PRR genes in murine skin in response to excisional injury. The genes are grouped by families: TLRs, NLRs, CLRs, and RLRs. Fold changes in transcript levels in wounded skin relative to levels in intact skin are presented. PRR genes with absolute fold changes >3.0 at at least one time point and FDR-adjusted P values below 0.05 (Benjamini–Hochberg procedure) are shown. (B) Time course of changes in Mincle protein levels in wounded murine skin were evaluated via Western blotting. (C) Time course of changes in Mincle protein levels in murine skin after the single topical application of 25 μL of 0.5% DNFB compared with the Mincle protein levels in the skin after vehicle application was evaluated via Western blotting. (D) Evaluation of Mincle expression in intact skin and in skin after wounding or single exposure of DNFB via immunostaining. The time point 2 h after skin wounding or DNFB exposure is shown. Shown are representative results obtained from skin sections of at least six animals in each group. (E) Fluorescent double staining of Mincle and dendritic cell markers in murine skin 2 h after a single exposure to 25 μL of 0.5% DNFB. Representative images are shown. (F) The secretion of proinflammatory mediators is significantly impaired in the absence of Mincle after skin wounding and after a single topical application of 25 μL of 0.5% DNFB. The level of the mediators was measured in the skin 12 h after wounding or DNFB or vehicle exposure. *P < 0.05 versus wild type (WT) (n = 5 mice per group; representative results from one of three independent experiments are shown). Mean values ± SD are shown.

To investigate changes in the expression of Clec4e (Mincle) upon skin injury at the protein level, we compared Mincle expression in damaged skin tissue at different stages of the wound repair process using Western blotting. In accordance with the gene array data, the expression of the Mincle protein increased quickly after injury and had decreased to baseline levels by 12 d after wounding (Fig. 1B). It is noteworthy that the rapid induction of Mincle expression was observed not only after the physical injury of the skin but also in response to the single topical application of the irritant and sensitizer DNFB onto the abdomen skin of mice (Fig. 1C). Immunohistochemical studies revealed a population of Mincle-expressing cells in the dermis that had mononuclear histiocyte-like morphological characteristics and that appeared in the damaged skin as an early response to wounding or DNFB application (Fig. 1D). Only a small number of weakly stained Mincle-expressing cells can be found in intact skin. Double immunofluorescent staining performed 2 h after DNFB application revealed that the Mincle-expressing cells do not express Langerin or CD11c in detectable quantities. However, these cells possess phenotype markers of plasmacytoid dendritic cells (pDC), such as BST2 (PDCA-1) and CD45R (B220) (Fig. 1E). Taken together, these findings indicate that Mincle is a receptor that is highly inducible in response to skin tissue damage.

To evaluate the role of Mincle in early inflammatory response, we measured concentration of proinflammatory mediators in skin at the early time point (12 h) after trauma using Mincle-deficient mice and wild-type mice. We determined that, after skin wounding or after a single topical application of DNFB, the early secretion of proinflammatory mediators in the absence of Mincle was significantly impaired (Fig. 1F). However, the absence of Mincle did not influence the dynamics of wound closure (Fig. S1).

Fig. S1.

Fig. S1.

Wound closure of full-thickness wounds in Mincle-deficient mice and wild-type mice. (A) Representative photographs of the wounds at different healing stages. (B) Reduction of wound areas at different time points expressed as a percentage of the initial wound size. Results are shown as mean ± SD (n = 5 mice per group; representative results from one of two independent experiments are shown).

The findings are consistent with those of recently reported studies that have shown increased Mincle expression in the brains of humans and rodents after brain injury (14, 15). These data, as well as the data presented here, allowed us to hypothesize that the Mincle may be an important sensor of nonphysiological cell death or cell stress that recognizes endogenous molecules released from damaged tissues and subsequently induces inflammation. The ribosomal protein SAP 130 has been reported to be an endogenous ligand for Mincle and to induce inflammation in vivo (7). We hypothesized that unknown endogenous lipid ligands for the Mincle receptor exist because the molecules of bacterial and fungal origin recognized by Mincle are amphiphilic lipids.

Isolation and Identification of Endogenous Mincle Ligands.

To evaluate whether Mincle recognizes endogenous lipids, we isolated the total lipid extract from mouse skin and tested its ability to induce the Mincle-dependent transcriptional activation of NF-κB in an NF-κB–driven luciferase reporter assay using a previously developed HEK293–mMincle–NF-κB–Luc cell line. As shown in Fig. 2A, the total lipid extract exhibited dose-dependent Mincle-stimulating activity. Next we performed a preliminary separation of the total lipid extract using a solid-phase extraction procedure. Using a reversed-phase cartridge, we found that the active compounds could be eluted from the cartridge almost without loss in their activating effect on Mincle (Fig. 2A). The chromatographic retention behavior suggested that the active components of the total lipid extract were amphiphilic molecules with both hydrophilic and hydrophobic fragments. Using electrospray ionization mass spectrometry, we found, perhaps expectedly, amphiphilic lipids in the eluate, particularly, cholesterol sulfate and phosphoinositols (Fig. S2). The extract obtained from the reversed-phase cartridge was further separated via HPLC in gradient separation mode using a reversed-phase monolithic column. Fractions were collected, and each fraction was evaluated using the reporter HEK293–mMincle–NF-κB–Luc cell line. The highest activity was detected in fraction 7 (Fig. 2B). One of the most intense signals in the mass spectrum (negative ion detection mode) of fraction 7 corresponded to a peak with an m/z of 465.31 (Fig. 2C). To elucidate the structure of this compound, this ion was isolated and subjected to fragmentation. Along with a peak corresponding to the parent ion in the tandem mass spectrum, only two product ion peaks were detected (m/z = 79.97 and m/z = 96.98, Fig. 2C). This finding suggests the presence of a sulfo-group in this compound and that the peak with an m/z of 465.31 corresponds to a deprotonated molecular ion [M-H]−. According to the LIPID MAPS database and MS-LAMP software, the compound was identified as a cholesterol sulfate. To confirm the identification, synthetic cholesterol sulfate was used as a standard. Its fragmentation spectrum completely matched the fragmentation spectrum of the ion from fraction 7 that had a peak with an m/z of 465.31 (Fig. 2E). Taken together, these data show that the main active component of fraction 7 was cholesterol sulfate.

Fig. 2.

Fig. 2.

Identification of a Mincle ligand of endogenous origin. (A) Serial dilutions of isolated total lipid extracts from murine skin samples or eluates from solid-phase extractions (reversed-phased C18 cartridge) were used to stimulate Mincle-expressing (HEK293–mMincle–NF-κB–Luc) or control (HEK293–NF-κB–Luc) reporter cell lines. TDB (2 µg/mL) and TNFa (100 ng/mL) were used as controls. Luciferase activity was measured and expressed as fold induction relative to the unstimulated control (K−). Mean values ± SD for triplicate measurements are shown. *P < 0.05 vs. control reporter cell line. Representative results from five independent experiments are shown. (B) Evaluation of the Mincle-mediated activity of fractions obtained via HPLC separation of eluates from solid-phase extractions using a Mincle-expressing reporter cell line. TDB (2 µg/mL) and TNFa (100 ng/mL) were used as controls. Luciferase activity was measured and expressed as fold induction relative to the unstimulated control (K−). Mean values ± SD for triplicate measurements are shown. Representative results from three independent experiments are shown. (C) Mass spectrum of fraction 7 in negative ion detection mode. (D) Fragmentation spectrum in negative ion mode of the ion with a m/z of 465.3113 from C. The proposed chemical structures of the parent ion and product ions are shown. (E) Fragmentation spectrum in negative ion mode of the synthetic cholesterol sulfate.

Fig. S2.

Fig. S2.

ESI-MS analysis of eluate after solid-phase extraction of skin lipids. (A) Mass spectrum in negative ion detection mode. The highest signals belong to cholesterol sulfate and phosphoinositols. (B) Chemical formulas of certain lipids identified based on their fragmentation spectra. Synthetic 1-stearoyl-2-arachidonoyl-sn-glycero-3-phosphoinositol, also called 18:0–20:4 phosphoinositol, was further used in binding studies to evaluate the specificity of the interaction of the Mincle receptor with cholesterol sulfate.

Mincle Recognizes Cholesterol Sulfate Through Direct Binding and Induces a Mincle-Dependent Proinflammatory Response Both in Vitro and in Vivo.

First we performed surface plasmon resonance experiments to study the ability of cholesterol sulfate to bind with Mincle using highly purified synthetic cholesterol sulfate. An anti-histidine antibody was first covalently immobilized onto the sensor chip surface, and the histidine-tagged recombinant Mincle protein was then injected and captured by the immobilized anti-histidine antibody, after which the analytes were injected. To evaluate the specificity of the interaction, we used a negative control, synthetic 18:0–20:4 phosphoinositol. The lipid was coeluted from a reversed-phase cartridge together with cholesterol sulfate during a solid-phase extraction of skin lipids (Fig. S2), and it shared some physicochemical properties with cholesterol sulfate. We observed the direct binding of Mincle to cholesterol sulfate but not to phosphoinositol. The obtained sensorgrams are presented in Fig. 3A.

Fig. 3.

Fig. 3.

Cholesterol sulfate directly binds to Mincle and induces Mincle-mediated proinflammatory signaling in vitro and in vivo. (A) Surface plasmon resonance analysis of the direct binding of Mincle to cholesterol sulfate. To verify binding specificity, 18:0–20:4 phosphoinositol was used. Shown are double-referenced sensorgrams (blank surface and blank buffer referencing) pertaining to the indicated concentrations of analytes. (B) Dose-dependent ability of cholesterol sulfate and TDB to activate Mincle-expressing reporter cells. Luciferase activity was measured and expressed as fold induction relative to the unstimulated control. Mean values ± SD for triplicate measurements are shown. *P < 0.05 vs. activations in the control reporter cell line. Representative results from five independent experiments are shown. (C) Cholesterol sulfate induces the secretion of proinflammatory mediators in a Mincle-dependent manner. BMDCs isolated from Mincle-KO mice or wild-type mice were stimulated with cholesterol sulfate (10 μg/mL). TDB (10 μg/mL) and LPS (10 μg/mL) were used as controls. Mean values ± SD for triplicate measurements are shown. *P < 0.05. Representative results from two independent experiments are shown. (D) Skin inflammatory response to a s.c. injection of cholesterol sulfate is mediated by Mincle. (Right) Higher magnification of areas indicated with arrows. Representative microphotographs of H&E-stained sections from one of three independent experiments are shown.

Next, we evaluated the ability of cholesterol sulfate to induce Mincle-dependent signaling using the reporter HEK293–mMincle–NF-κB–Luc cell line. The reporter cells showed a similar response to both cholesterol sulfate and trehalose dibehenate, an archetypical ligand of Mincle (Fig. 3B). Thus, this finding indicates that cholesterol sulfate binds to Mincle and induces Mincle-mediated signaling.

To determine the specificity of cholesterol sulfate signaling through Mincle under more physiologically realistic conditions and to study the relevance of the cholesterol sulfate/Mincle interaction in vivo, we used Mincle-deficient mice. To compare the ability of cholesterol sulfate and trehalose dibehenate to induce cytokine and chemokine secretion, bone marrow-derived dendritic cells (BMDCs) were isolated from Mincle-KO mice and wild-type mice and stimulated by these lipids. The levels of cytokines and chemokines in culture supernatants were measured using a multiplex immunoassay. Both cholesterol sulfate and trehalose dibehenate induced the secretion of proinflammatory mediators, such as IL-1a and IL-1b, KC, and MIP-1a and MIP-1b (Fig. 3C). The secretion of these proinflammatory mediators in response to both cholesterol sulfate and trehalose dibehenate was significantly reduced in BMDCs from Mincle-deficient mice, whereas the response to bacterial lipopolysaccharide (an agonist of TLR4) remained unchanged (Fig. 3C).

To evaluate the tissue response to cholesterol sulfate and the contribution of Mincle to this process, we subcutaneously injected a sterile aqueous solution containing micelles of cholesterol sulfate into either Mincle-deficient or wild-type mice. Sterols tend to form crystals in solution, and these crystals are potent activators of inflammatory responses (16, 17). To avoid the injection of a crystal-containing suspension, we previously developed a method using an ultrasonic treatment to produce a stable micelle solution with an average micelle diameter from 70 to 90 nm (Fig. S3). After 24 h, skin samples were excised and the local tissue response was evaluated based on a histological analysis of hematoxylin and eosin (H&E)-stained sections. The histological examination revealed a marked inflammatory infiltrate in the skin of wild-type animals injected with cholesterol sulfate, whereas, in Mincle-deficient animals, the accumulation of infiltrating cells was significantly reduced (Fig. 3D). The infiltrating cells were primarily neutrophils, monocytes, and eosinophils. Taken together, our findings suggest that a Mincle deficiency results in a reduced inflammatory response to cholesterol sulfate both in vitro and in vivo.

Fig. S3.

Fig. S3.

Ultrasonic treatment produced a stable solution of micelles of cholesterol sulfate, which was suitable for use in the in vivo experiments. The size distribution of cholesterol sulfate micelles (A) immediately after ultrasound treatment and (B) after 1 h.

Mincle Promotes Cutaneous Allergic Inflammation.

Taking into account (i) the significant inducibility of Mincle expression in skin to the topical skin irritation caused by DNFB, a well-known inductor of allergic skin reactions, and (ii) the significantly impaired secretion of proinflammatory mediators in response to a single DNFB application in Mincle-deficient animals, we examined the role of Mincle in the pathogenesis of cutaneous allergic inflammation. To address the role of Mincle in the development of cutaneous allergic inflammation, we used one of the most commonly used models of contact hypersensitivity (CHS) that adequately reflect allergic contact dermatitis (18). Mincle-KO and wild-type mice were sensitized to DNFB on their abdomens, followed by challenges with the same sensitizer on the dorsum of both ears. The schedule for the mouse sensitization and challenge procedure is depicted in Fig. 4A. Endpoint parameters were measured 24 h after the last DNFB exposure. Under the induction protocol, Mincle-deficient mice exhibited significantly suppressed clinical symptoms of allergic contact dermatitis, such as redness and ear swelling, compared with the wild-type animals (Fig. 4 B and C). A careful review of H&E-stained stained sections of contact-allergic ear tissue at the experiment endpoint revealed a strongly reduced inflammatory response in Mincle-deficient mice in terms of ear thickness, epidermal acanthosis, and inflammatory cell infiltrate (Fig. 4D). A quantitative analysis using flow cytometry confirmed that there were significantly fewer infiltrating Gr1+/CD11b+ myeloid cells in the ear tissue of Mincle-deficient mice compared with wild-type control mice (Fig. 4E). To evaluate the differences in allergic skin inflammation at the molecular level, we measured the production of cytokines and chemokines in inflamed ear tissues. We found that a Mincle deficiency led to the suppressed production of a spectrum of mediators of inflammation (Fig. 4F), including proinflammatory cytokines IL-1b, TNFa, and IL12p40, as well as chemokines KC, MIP-1a, and MIP-1b. Interestingly, the production of IL-17A was also significantly suppressed in the inflamed tissues of Mincle-deficient mice compared with IL-17A’s production in wild-type animals (Fig. 4F). Taken together, these observations demonstrate that Mincle strongly enhances the magnitude of the skin inflammatory CHS response and plays a role as a driving component in the pathogenesis of allergic skin inflammation.

Fig. 4.

Fig. 4.

Mincle promotes allergic skin inflammation in a model of allergic contact dermatitis. Mincle-KO and wild-type mice were sensitized to DNFB on their abdomens, followed by challenges with the same sensitizer on the dorsum of both ears. Endpoint parameters were measured 24 h after the last DNFB exposure. As a negative control, two other groups of Mincle-KO and wild-type mice were exposed to the vehicle without DNFB throughout the duration of experiment. *P < 0.05 versus WT (n = 6 mice per group; combined data of three independent experiments). Mean values ± SD are shown. (A) The schedule for mouse sensitization and challenges is shown. C, challenge; E, experimental endpoint; S, sensitization. (B) Dynamics of changes in ear thickness. (C) Representative photographs of mouse ears at the experimental endpoint are shown. The photographs illustrate decreases in certain clinical symptoms of allergic contact dermatitis, such as ear swelling and redness, in Mincle-KO mice. (D) Representative H&E-stained sections of ear tissue in wild-type and Mincle-KO mice at the experimental endpoint are shown. (E) Recruitment of Gr1+/CD11b+ myeloid immune cells in ear tissue at the experimental endpoint as assessed via flow cytometry (Left) and presented as percentages of cells (Right). (F) Analysis of cytokine and chemokine production in mouse ear tissue at the experimental endpoint.

Discussion

There is growing evidence that the PRRs of the innate immune system play a key role in the induction of rapid inflammation in response to infection and tissue trauma (3, 19, 20). The description of PRRs as a limited number of evolutionarily conserved receptors that recognize molecular patterns of infectious agents, the so-called PAMPs, was first developed by Janeway (21, 22). Initially, PRRs were considered as a first line of host defense during an infection, functioning in discriminating between self and nonself. Later, Matzinger proposed the existence of host molecules (DAMPs) that are released or secreted in response to tissue damage and can initiate and perpetuate an immune response (23, 24). Subsequently, it was proven that one of the functions of PRRs is the sensing of tissue damage, and many DAMPs had been identified currently (13, 19). Recently, Medzhitov et al. proposed that allergic reactions are a component of the host defense system against noninfectious noxious environmental factors, including venoms, noxious xenobiotics, and irritants that can induce tissue damage (25). However, despite extensive research in recent years, our understanding of the molecular mechanisms underlying the physiological inflammatory response to trauma and pathological sterile inflammation remains incomplete.

This study shows that the pattern recognition receptor Mincle (Clec4e), which is highly inducible in skin in response to injury and irritation, (i) recognizes cholesterol sulfate and subsequently induces a proinflammatory response and (ii) plays a role as a driving component in the pathogenesis of allergic skin inflammation.

Previously, it was reported that sterols are able to cause inflammation due to their interaction with the innate immune system, and some mechanisms for this effect were proposed (16, 17). The ability of cholesterol to bind to Mincle has recently been shown; however, the ability of cholesterol to cause Mincle-mediated inflammation in vivo was not demonstrated (26). Here, we show that cholesterol sulfate is an endogenous ligand for Mincle, and we studied the inflammatory response induced by the cholesterol sulfate/Mincle interaction in detail both in vitro and in vivo. Mincle recognizes cholesterol sulfate through direct binding and subsequently induces the secretion of a range of proinflammatory mediators, such as IL-1a, IL-1b, KC, MIP-1a, and MIP-1b. In the in vivo experiments, the s.c. injection of cholesterol sulfate resulted in a Mincle-mediated induction of a severe local inflammatory response. The effects were significantly reduced when Mincle signaling was absent. However, some residual proinflammatory activity of cholesterol sulfate in the absence of Mincle was detected, suggesting the presence of compensatory mechanisms. It is possible that another CLR, MCL (also known as Clec4d or Dectin-3), is involved in the compensatory mechanisms. Mincle and MCL are homologous, and they are located next to each other in the genome, suggesting that MCL may have originated from a gene duplication of Mincle (13). Both MCL and Mincle recognize TDM and use the same FcRγ-Syk-CARD9 pathway (13, 27). We revealed that MCL and Mincle are extremely inducible in skin after trauma (Fig. 1A). Using the reporter HEK293–mMCL–NF-κB cell line, we found that cholesterol sulfate is able to induce MCL-mediated signaling (Fig. S4). However, additional studies are required to investigate the consequences of the interaction between cholesterol sulfate and MCL in more detail.

Fig. S4.

Fig. S4.

Dose-dependent ability of cholesterol sulfate to activate HEK–mMCL–NF-κB–Luc reporter cells expressing MCL (Clec4d). TDB (2 µg/mL) and TNFa (100 ng/mL) were used as controls. Luciferase activity was measured and expressed as fold induction relative to the unstimulated control (K−). Mean values ± SD for triplicate measurements are shown. *P < 0.05 vs. activation of the control reporter cells. Representative results from five independent experiments are shown.

The post-traumatic inflammatory response is generally considered a first step in tissue repair (5). However, the signaling through Mincle induced by endogenous stimuli may also underlie the pathogenesis of certain diseases associated with sterile chronic inflammation. Recently published results indicate that signaling through Mincle plays a role in obesity-induced adipose tissue inflammation and the subsequent adipose tissue fibrosis (28). Recently, an essential role of Mincle signaling through the Syk/Card9 axis in the development of autoimmune eye inflammation in a model of experimental autoimmune uveoretinitis was revealed (29). It was shown that Mincle signaling promotes pancreatic oncogenesis, whereas the deletion of Mincle protects against oncogenesis (30). Interestingly, T cells, which are not protective against the progression of pancreatic ductal adenocarcinoma in mice with intact Mincle signaling, are reprogrammed into indispensable mediators of antitumor immunity in the absence of Mincle (30).

Here, we show the driving role of Mincle in the development of cutaneous allergic inflammation. Using a classical model of allergic contact dermatitis, we show that Mincle deficiency strongly suppresses the magnitude of the skin inflammatory CHS response, as reflected by changes in ear swelling, myeloid cell infiltration, and cytokine and chemokine production in inflamed tissues.

It was previously shown that the application of contact allergens leads to the activation of mechanisms related to innate immunity through signaling pathways that are also involved in antiinfectious immunity (31). Interestingly, it has been demonstrated that the strength of primary skin inflammation in response to contact allergens determines the strength of the subsequent CHS response and whether a response or tolerance develops (3134). Expression of Mincle is rapidly induced in skin as a response to wounding or a DNFB single application in the cells that bear specific pDCs markers [CD45R (B220) and, importantly, BST2 (PDCA-1), which is considered to be a unique marker of pDCs]. It was previously demonstrated that pDCs are resident dendritic cells that normally occur in the dermis only rarely but can accumulate in lesioned skin (35). We have shown that the early secretion of proinflammatory mediators in response to wounding or to a DNFB single application in the absence of Mincle was significantly impaired. Mincle uses the Syk/CARD9-signaling pathway, and the pathway in dendritic cells was recently shown to be a crucial signaling axis in induction of the skin CHS response (36). Therefore, endogenous Mincle ligands may serve as auto-adjuvants, activating the innate immune system. Interestingly, the activation of Mincle by its ligands, such as trehalose-6,6′-dimycolate from M. tuberculosis or the synthetic adjuvant trehalose dibehenate, promotes Th1/Th17 T-cell responses (9, 13, 37). We found in allergic inflamed ears from Mincle-KO mice a suppressed production of IL-17A, which is regarded as a signature cytokine for Th17 cells (38). Th17 cells play a crucial role as effector cells in the pathogenesis of allergic contact dermatitis, psoriasis, and autoimmune diseases (3941). Thus, the activation of Mincle during the development of allergic contact dermatitis may lead to a bias that influences T-cell polarization. However, further studies are needed to determine the precise mechanism underlying the driving role of Mincle in cutaneous allergic inflammation.

Cholesterol sulfate is present in various tissues and body fluids, and it accumulates during squamous cell differentiation in the epithelial cells of barrier tissues, such as the respiratory tract and esophageal mucosa (11, 12, 42). Cholesterol sulfate is also found in considerable quantities in the skin, predominantly in the epidermis (10), where it acts as an important regulator during the formation of the epidermal barrier (42).

Based on our findings, it can be hypothesized that Mincle, the expression of which is rapidly increased in dermis in response to skin trauma or irritation, can recognize cholesterol sulfate released from the damaged epidermis and subsequently amplify an initial inflammatory response in the skin to activate and mobilize skin antigen-presenting cells. Although DNFB application onto the abdominal skin of mice does not lead to significant changes in the quantity of cholesterol sulfate per skin biopsy sample (Fig. S5), it could be hypothesized that the DNFB application leads to rapid cholesterol sulfate permeation from epidermis to dermis. Also, it could be hypothesized that the activity of CD8+ cytotoxic T cells during the challenge phase that causes epithelial cell damage may also lead to the release of cholesterol sulfate and subsequently Mincle activation. However, it is possible that other endogenous ligands of Mincle, such as SAP130, may play an important role in the Mincle activation during the skin inflammatory response. Additional investigations are necessary to precisely determine the role of cholesterol sulfate/Mincle interaction in the pathogenesis of allergic skin inflammation.

Fig. S5.

Fig. S5.

Quantification of cholesterol sulfate in intact skin and in skin after single exposure to DNFB or vehicle application. Using an 8-mm biopsy punch instrument, skin samples were collected and then weighed. After homogenization and lipid extraction, the quantity of cholesterol sulfate was determined using high-performance liquid chromatography with mass spectrometry detection. The combined data of two independent experiments are presented. Mean values ± SD are listed (n = 10 mice per group). DNFB application does not lead to statistically significant changes in the quantity of cholesterol sulfate per skin biopsy sample. The observed decrease in the cholesterol sulfate concentration per gram of wet tissue is likely due to skin swelling and the increased weight of the biopsy samples.

Taken together, our results demonstrate an important role of Mincle as a driving component in the pathogenesis of allergic skin inflammation and as a receptor of a DAMP, cholesterol sulfate. These results contribute to a deeper understanding of the fundamental innate immune mechanisms underlying sterile inflammation.

Materials and Methods

Male BALB/c mice and C57BL/6 and C57BL/6-Mincle-KO mice of both sexes were used in this study. All mice were between 9 wk and 10 wk of age. The Mincle-KO knockout mouse line was obtained from the National Institutes of Health-sponsored Mutant Mouse Regional Resource Center (MMRRC) National System and was back-crossed with C57BL/6 mice for 10 generations. All of the animal experimental procedures were in line with the Bioethics Commitee of N. F. Gamaleya Federal Research Center of Epidemiology and Microbiology guidelines.

RNA samples for microarray analysis were prepared as described previously (43, 44). Total lipids were isolated from skin samples using sequential extractions with different solvent mixtures according to a previously described procedure (45). Lipids were then fractionated according to a previously reported procedure (46). Separation via HPLC was achieved as suggested by Ikeda et al. (47) with some changes. BMDCs from Mincle-KO and wild-type mice were differentiated from proliferating mouse bone marrow progenitors according to a previously described protocol (48). A well-known murine model of allergic contact dermatitis was induced using a method described previously (18). Cholesterol sulfate quantification in skin was performed according to a protocol (49) with some modifications.

Additional information is provided in SI Materials and Methods.

SI Materials and Methods

Mice.

Male BALB/c mice (Stolbovaya Nursery of the Russian Academy of Medical Science) and C57BL/6 and C57BL/6-Mincle-KO mice of both sexes were used in this study. All mice were between 9 wk and 10 wk of age. The Mincle-KO knockout mouse line was obtained from the National Institutes of Health-sponsored MMRRC National System and was back-crossed with C57BL/6 mice background for 10 generations. The mice were fed a completely pelleted laboratory chow and had access to food and water ad libitum.

Wounding Procedure.

Male BALB/c mice (for microarray analysis) or male C57BL/6 and C57BL/6-Mincle-KO mice were used. The mice were anesthetized intraperitoneally with xylazine (7.5 mg/kg) and Zoletil (45 mg/kg, Virbac). The backs of the mice were shaved with an animal clipper and disinfected with 70% (vol/vol) ethanol. Two full-thickness dermal wounds were made on opposite sides of the midline of each mouse using a 4-mm punch biopsy instrument (Dermo-Punch). Wounds were made through the epidermis, dermis, and s.c. tissue layer, leaving the fascia intact. The animals were individually caged after wounding. At a specified time point after wounding, mice were killed via carbon dioxide inhalation. Then the wounds and surrounding tissues or intact skin samples were removed with an 8-mm biopsy punch.

In Vivo Wound-Healing Assay.

Digital photographs of the wounds were taken on days 1, 3, 7, and 12 after wounding. The area of the wound was then measured on the photographs in pixels using Adobe Photoshop CS6 (Adobe).

Microarray Analysis.

Tissue samples were crushed in liquid nitrogen. QIAzol lysis reagent (Qiagen) was added, and the homogenate was centrifuged through a QIAshredder column (Qiagen). Total RNA was extracted from the eluate using the miRNeasy mini kit (Qiagen) according to the manufacturer’s instructions. Genomic DNA contamination was removed by performing a DNaseI digestion on an RNA-binding column for 15 min. Total RNA was eluted in 50 μL of RNase-free water and stored at −80 °C. RNA yield and purity were assessed spectrophotometrically by measuring OD260 and the OD260/280 ratio, respectively, in RNase-free water using a NanoDrop ND-1000 (NanoDrop Technologies). For all samples, the OD260/280 ratio was between 2.0 and 2.2.

RNA integrity was determined using an Agilent Bioanalyzer 2100 system (Agilent Technologies). RNA quality indicator (RIN, RNA integrity number) values generated by the Bioanalyzer system’s software ranged from 7.7 to 9.1.

RNA samples were prepared according to the manufacturer’s instructions (Affymetrix Manual P/N 701880 Rev. 4) using 500 ng of total RNA as the starting material as described previously (43, 44). The samples were hybridized on GeneChip Mouse Gene 1.0 ST Arrays (Affymetrix) for 16 h at 45 °C. Arrays were washed to remove nonspecifically bound nucleic acids and stained on a Fluidics Station 450 (Affymetrix) using the FS450_0007 protocol followed by scanning on a GeneChip Scanner 3000 7G system (Affymetrix). The microarray CEL files have been deposited in the GEO database (accession no. GSE76144).

Arrays were preprocessed in GeneChip Command Console Software (AGCC, Affymetrix, version 1.4.1.46) and the Affymetrix Transcriptome Analysis Console (version 3.0.0.466) using the RMA-sketch option. Differentially expressed genes were determined based on a moderated t test using the limma package from the Bioconductor project (www.bioconductor.org). Genes with absolute fold changes above 3.0 and FDR-adjusted P values below 0.05 (Benjamini–Hochberg procedure) were selected.

Western Blot Analysis.

Skin samples containing wounds or sites of DNFB or vehicle application as well as intact skin were removed using an 8-mm biopsy punch and then immediately placed into ice-cold T-PER extraction buffer (T-PER, Pierce) containing a complete protease inhibitor (Roche Diagnostics). Approximately 0.75 mL of the buffer was used per 35 mg of skin sample. The homogenized samples were prepared using a FastPrep 24 device and tubes containing lysing matrix A (MP Biomedicals). The homogenates were centrifuged at 12,000 × g for 12 min at 4 °C. The total protein in the supernatants was measured using the Bradford method, and protein levels in the supernatants were then normalized. Next the protein extracts were separated via SDS/PAGE, transferred onto nitrocellulose membranes, and probed with antibodies against Clec4e (1:1,000, D292-3, MBL International) or β-actin (1:3,000, ab8227, Abcam). Proteins of interest were detected with an HRP-conjugated goat anti-rat IgG antibody (1:10,000, NA935, GE Healthcare) or goat anti-rabbit IgG antibody (1:2,000, ab6721, Abcam) and visualized using the Optiblot ECL detection kit (ab133406, Abcam) according to the provided protocol.

Immunohistochemistry.

Skin samples containing wounds or sites of DNFB or vehicle application as well as intact skin were removed using an 8-mm biopsy punch and frozen in liquid nitrogen. Next we cut 4-μm sections of the skin samples and fixed them using 4% (vol/vol) paraformaldehyde. The samples were blocked with BSA (Sigma-Aldrich) for 30 min at 37 °C. They were then incubated with primary antibodies for 1 h at 37 °C. The following primary antibodies were used: anti-Clec4e (1:200, orb1341, Biorbyt), anti-CD45R/B220 (1:200, 103201, BioLegend), anti-BST2 (120G8.04) (1:10, DDX0390P, Novus Bio), anti-CD11c (117302, BioLegend), and anti–Langerin-PE (1:200, 144204, BioLegend). After washing, we incubated the samples with secondary antibodies for 30 min at 37 °C. Next, the cell nuclei were costained with DAPI (Sigma-Aldrich). The following secondary antibodies conjugated with fluorochromes were used: anti-rabbit antibodies (1:100, 711–585-152, Jackson ImmunoResearch), anti-rat antibodies (1:500, 10123952, Invitrogen), and anti-Armenian hamster antibody (1:50, 405512, BioLegend). The samples were photographed using a Keyence microscope (Model BZ-9000) and a confocal microscope (TCS SP5 STED; Leica Microsystems).

Extraction of Total Lipids from Skin.

Intact murine skin samples were collected, weighed, cut with scissors, and then immediately placed into ice-cold RIPA buffer. Approximately 7.5 mL of the buffer was used per 1 g of skin. The homogenized samples were prepared using a FastPrep 24 device and tubes containing lysing matrix A (MP Biomedicals). Total lipids were isolated from the obtained homogenate using sequential extractions with different solvent mixtures according to a procedure described previously (45). The obtained total lipid extract was dried under a vacuum.

Solid-Phase Extraction.

Approximately 25 mg of the obtained total lipid extract was emulsified in 10 mL of a chloroform–methanol–water (3/48/47) mixture containing 0.1 M NaCl, and the emulsion was then sonicated using an ultrasound bath. The lipids were fractionated according to a previously reported procedure (46). Briefly, the lipid extract was applied to a pre-equilibrated Sep-Pak C18 cartridge (WAT051910, Waters) connected to an AKTA Explorer 10 chromatography system (GE Healthcare), and the cartridge was then sequentially washed at a flow rate of 3 mL/min with a chloroform–methanol–water (3/48/47) mixture containing 0.1 M NaCl, followed by water and, finally, methanol. The methanol-eluted fraction was then collected and dried using a vacuum rotary evaporator.

Separation via HPLC.

HPLC separations were achieved as described by Ikeda et al. (47) with several important changes. Briefly, HPLC was performed using a monolithic reversed-phase Chromolith Performance RP-18e column (4.6 mm × 100 mm, Merck). Separations were performed using a Waters BioAlliance 2796 HPLC module (Waters). The column temperature was held at 40 °C. The mobile phases were prepared from mixtures of methanol (A), isopropanol (B), and water (containing 200 mM ammonium formate) (C). The gradient consisted of holding solvent D (A/B/C, 55/25/20 by volume) for 5 min, linearly converting solvent D to solvent E (A/B, 60/40 by volume) over 10 min, and then holding solvent E for 5 min. After a run was completed, the gradient was returned to solvent D for over 5 min, and before the next run, an equilibration of the column was performed for 5 min. The mobile phase was pumped at a flow rate of 1.5 mL/min for gradient elution. Approximately 1 mg of obtained, dried eluate from the solid-phase extraction was dissolved in solvent D. After sonication in an ultrasound bath, 100 μL of the sample solution was applied to the column, and 1-mL fractions were collected using a fraction collector. Fractions of the same number from three independent runs were combined and then dried using a vacuum rotary evaporator.

Lipid Identification via Electrospray Ionization Mass Spectrometry Analysis.

The electrospray ionization mass spectrometry (ESI-MS) analyses were performed using a QSTAR Elite system (AB Sciex) equipped with a TurboIonSpray ion source via direct infusion using a built-in Harvard Syringe Pump at a flow rate of 5 µL/min. Full-scan profiling data were acquired in the TOF mass analyzer in positive and negative modes, and the source and ion transfer parameters applied were as follows: ion source nebulizer gas (GS1), 3 L/min; ion source heater gas (GS2), 3 L/min; gas temperature (TEM), 300 °C; and curtain gas flow rate (CUR), 20 L/min. The ionspray voltage (IS) was −4,500 V in negative mode and 5,500 V in positive mode. The declustering potential (DP) was −70 V, and the focusing potential (FP) was −40 V. Tandem mass-spectrometry experiments (MS/MS) were performed using the same parameters for the ion source and optics as described above. Parent ions were isolated in quadrupole Q1 in the “Unit Resolution” mode. The collision energy for ion fragmentation was set to 40 eV.

An identification search was performed in the LIPID MAPS database (www.lipidmaps.org) with the help of MS-LAMP software (ms-lamp.igib.res.in) using the following queried parameters: m/z, 465.3113 (monoisotopic); ion type, [M-H]−; window range, ±0.01; class, sulfur-containing lipids. As a result of this query, only one possible molecule, cholesterol sulfate, was proposed; thus, the precision of the mass measurement was 0.0074 Da or 16 ppm.

Synthetic cholesterol 3-sulfate (700016P, Avanti Polar Lipids) was used as a standard.

Lipid Preparation.

A micelle-containing solution of cholesterol 3-sulfate (700016P, Avanti Polar Lipids) was prepared at a concentration of 1 mg/mL for in vivo treatments using a Branson S-450D instrument (Branson). Endotoxin-free water was obtained from a Milli-Q Advantage A10 system (Millipore). The ultrasonic treatment produced a solution of micelles that were stable for at least 1 h (according to the size and size distribution measurements, Fig. S3), with the average micelle diameter ranging from 70 nm to 90 nm. Before injection, the solution was sterilized via filtration through a 22-µm filter. Measurements of cholesterol sulfate micelle size and size distribution were performed in triplicate using a Zetasizer Nano ZS (ZEN3600, Malvern Instruments Ltd.) at 25 °C. All calculations based on light-scattering intensity were performed using Zetasizer Nano ZS software.

To perform the Surface Plasmon Resonance (SPR) experiments and the reporter cell-line assays, synthetic lipids (trehalose dibehenate, 890808; cholesterol sulfate, 700016P; 18:0–20:4 phosphoinositol, 850144P; all from Avanti Polar Lipids) as well as dried total lipid extract aliquots and separated fractions were solubilized using the ultrasound treatment described above. Endotoxin-free water was used as a solvent in the reporter cell-line assays, and binding buffer was used in the SPR experiments. Then serial dilutions were performed, and each dilution was sonicated.

For experiments with BMDCs, 1 mg of cholesterol sulfate (700016P, Avanti Polar Lipids) or trehalose dibehenate (vac-tdb, InvivoGen) was dissolved in 100 µL of DMSO (DMSO Hybri-Max, Sigma-Aldrich) and heated at 60 °C for 30 s. Then 900 µL of water was added, followed by heating for 15 min at 60 °C, after which serial dilutions were performed. In in vitro experiments, bacterial lipopolysaccharide from Escherichia coli O111:B4 (tlrl-pelps, InvivoGen) and recombinant human TNFa (rhtnf-a, InvivoGen) were used as controls.

SPR-Binding Experiment.

The SPR experiments were performed using a BIACORE 3000 (GE Healthcare) equipped with a research-grade CM5 sensor chip (BR100012, GE Healthcare) at a temperature of 25 °C. For binding analyses, the anti-histidine antibody from the His Capture Kit (28-9950-56, GE Healthcare) was first covalently immobilized onto the sensor chip surface at a level of ∼15,000 response units (RU) using the Amine Coupling Kit (BR-1000-50). Histidine-tagged recombinant human Clec4e, derived from human cells (C588, Novoprotein) in running buffer (25 mM Tris⋅HCl, 150 mM NaCl, 2 mM CaCl2, pH 7.2), was then injected over flow cell 2 (Fc2) at a concentration of 10 μg/mL at a flow rate of 15 µL/min and captured by the immobilized anti-histidine antibody. Flow cell 1 (Fc1), with immobilized anti-histidine antibodies, was intact and used as a blank surface. The analytes cholesterol 3-sulfate, sodium salt (700016P, Avanti Polar Lipids), and 18:0–20:4 phosphoinositol (1-stearoyl-2-arachidonoyl-sn-glycero-3-phosphoinositol, ammonium salt, 850144P, Avanti Polar Lipids) were prepared in running buffer using the ultrasound treatment described above. The analytes were injected over the two flow cells (Fc1 and Fc2) at a flow rate of 20 μL/min, and the complex was then allowed to associate and dissociate. Then the surfaces were regenerated with a 30-s injection of the regeneration solution (10 mM Tris⋅glycine, pH 1.5). During regeneration, the captured histidine-tagged ligand and the associated analyte were removed from the sensor chip surfaces, making it ready for the next cycle. Injections of each analyte concentration and a buffer blank were flowed over the two flow cells. Data were collected at a rate of 1 Hz. The data were double-referenced (blank surface and blank buffer referencing) using BIAevaluation software (GE Healthcare).

Reporter Cell-Line Preparation.

Mouse Clec4e (mMincle, gene ID 56619) and mouse Clec4d (mMCL, gene ID 17474) cDNA sequences were synthesized by Evrogen and inserted into pAI-TA2 vectors (Evrogen). For lentiviral stock preparation, Clec4e and Clec4d gene nucleotide sequences were cloned in the lentiviral plasmids pLV-Neo and pLV-Bleo, respectively. For the preparation of lentiviral particles, 293T cells (ATCC, CRL-3216) were transfected with vectors coding for Clec4e (pLV-Clec4e-Neo) or Clec4d (pLV-Clec4d-Bleo), together with the NF-κB–Luc construct (pLA-NF-κB–Luc), using Lipofectamine reagent (Invitrogen) according to the manufacturer’s instructions. Two helper plasmids, pCMVΔR8.2 and pVSV-G, were cotransfected along with the experimental vector. Supernatants containing infectious viral particles were harvested 24, 36, and 48 h post transfection, pooled, and filtered through a 0.22-µm filter.

HEK293 cells (ATCC, CRL-1573) were transduced with the lentiviral vector containing the reporter NF-κB–Luc construct pLA-NF-κB–Luc and were then cultured in the presence of 1 µg/mL puromycin (Invitrogen) for 2 d to produce a control reporter cell line (HEK293–NF-κB–Luc). Next, the control cells were transduced with either Mincle- or MCL-coding lentiviral vectors and cultured in 500 µg/mL G418 sulfate and 10 µg/mL bleomycin (both from Invitrogen) for 1 wk to produce stable cell lines (HEK–mMincle–NF-κB–Luc and HEK–mMCL–NF-κB–Luc). The expression of Mincle and MCL in the reporter cells was validated via Western blotting using anti-Mincle antibodies (1:1,000, D292-3, MBL International) or anti-MCL antibodies (1:200, sc-134743, Santa Cruz Biotechnology). Functional testing was performed by assessing the nuclear translocation of NF-κB after stimulating the cells with trehalose dibehenate. Western blotting of nuclear fractions was then performed using anti-p65 antibodies (1:2,000, ab16502, Abcam) as described elsewhere.

The reporter cells were maintained in DMEM (PAA Laboratories) supplemented with 10% (vol/vol) FCS (Thermo Scientific), 50 U/mL penicillin, 50 μg/mL streptomycin, 2 mM glutamine, and 0.1 M NaHCO3 (all from PanEco) at 37 °C with 5% (vol/vol) CO2.

Reporter Cell-Line Assay.

HEK–mMincle–NF-κB–Luc or HEK–NF-κB–Luc cells were seeded at 2 × 104 cells/well in 96-well plates (100 µL/well). Cells were treated with the substances of interest for 8 h. After incubation, the cells were lysed by adding 100 µL per well of GloLysis buffer (Promega) according to the manufacturer’s instructions. Luciferase activity was measured in the cell lysates using the Bright-Glo luciferase assay system (Promega). Luminescence was measured in relative units using a Synergy H4 hybrid reader (Bio-Tek Instruments).

BMDC Preparation.

BMDCs from C57BL/6 and C57BL/6-Mincle-KO knockout mice were differentiated from proliferating mouse bone marrow progenitor cells via induction with 20 ng/mL of granulocyte macrophage colony-stimulating factor (GM-CSF) (R&D Systems) over 6–9 d as described previously (48). Briefly, mice were euthanized with carbon dioxide. Their femurs and tibias were collected in ice-cold HBSS (HBSS, Sigma-Aldrich). The muscles were removed with a scalpel and by rubbing the bones with a paper tissue. The ends of the bones were then cut off with scissors and crushed. The bone marrow was flushed out with 2–3 mL of RPMI complete medium in a syringe with a 25-gauge needle. All bone marrow cells were collected and washed twice with HBSS. The bone marrow cells were cultured in 24-well plates containing ∼5 × 105 cells/mL in 1 mL total volume. The cells were maintained at 37 °C with 5% (vol/vol) CO2 in complete RPMI medium with 10% (vol/vol) heat-inactivated FCS (PAA Laboratories), 0.05 M mercaptoethanol (Thermo Fisher Scientific), nonessential amino acids (PanEco), 20 ng/mL GM-CSF, 2 mM glutamine, 100 U/mL penicillin, and 100 g/mL streptomycin (all PanEco). After 3 d, the nonadherent cells were collected and discarded and 1 mL of fresh medium was added to each well. On day 5, half of the medium was replaced with fresh medium. On day 7, the cells were collected, washed, and resuspended in medium without GM-CSF. After 7–9 d of cultivation, the percentage of CD11c+ cells was ∼60–70%. BMDCs obtained from C57BL/6 and C57BL/6-Mincle-KO knockout mice were seeded in 96-well plates at 1 × 105 cells per well (200 µL/well) in complete RPMI medium. Cells were treated with the substances of interest for 48 h. Plates were then centrifuged at 1,000 × g for 10 min, and the culture supernatants were collected.

In Vivo Cholesterol Sulfate Treatments.

C57BL/6 and C57BL/6-Mincle-KO knockout mice were anesthetized intraperitoneally with xylazine (7.5 mg/kg) and Zoletil (45 mg/kg, Virbac), and the backs of the mice were shaved with an animal clipper. After disinfection with 70% (vol/vol) ethanol, mice were injected s.c. with 20 µL of cholesterol sulfate solution or just the solvent (sterile water) as a negative control. Twenty-four hours post injection, the mice were anesthetized, and skin samples from the injection site were removed with an 8-mm biopsy punch.

Induction of Allergic Contact Dermatitis.

Allergic contact dermatitis was induced using a method described previously (18) with minor modifications. Briefly, Mincle-KO and wild-type mice were sensitized with 25 μL of 0.5% DNFB (Sigma-Aldrich) on the shaved abdomen for 2 consecutive days (days 0 and 1). Immediately before application DNFB was dissolved in vehicle (acetone (AppliChem) and olive oil (Sigma-Aldrich) in a proportion of 4:1 by volume). Challenges with 10 μL of 0.3% DNFB were performed on the dorsum of both ears on days 5, 6, 7, and 8, and the endpoint parameters were measured 24 h after the last DNFB exposure. As a negative control, two other groups of Mincle-KO and wild-type mice were exposed to the vehicle without DNFB for the duration of the experiment. Ear thickness was measured with a digital micrometer (Mitutoyo).

Cytokine and Chemokine Assays.

Skin samples containing wounds or sites of DNFB or vehicle application as well as intact skin were removed using an 8-mm biopsy punch. The skin samples or ears were immediately placed into ice-cold T-PER extraction buffer (T-PER, Pierce) containing a complete protease inhibitor (Roche Diagnostics). Approximately 0.75 mL of the buffer was used for each skin sample or ear. The homogenized samples were prepared using a FastPrep 24 device and tubes containing lysing matrix A (MP Biomedicals). The homogenates were centrifuged at 12,000 × g for 12 min at 4 °C. The total protein in the supernatant was measured using the Bradford method.

Levels of 23 cytokines and chemokines [IL-1α, IL-1β, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12 (p40), IL-12 (p70), IL-13, IL-17A, eotaxin (CCL11), G-CSF, GM-CSF, IFN-γ, KC (CXCL1), MCP-1 (CCL2), MIP-1α (CCL3), MIP-1β (CCL4), RANTES (CCL5), TNF-α] were measured in triplicate in the prepared cell culture supernatants and in the tissue homogenates using a 23-plex ELISA Bio-Plex Pro kit (Bio-Rad Laboratories) according to the manufacturer’s instructions. The concentrations of some selected cytokines and chemokines in cell culture supernatants were validated using commercial ELISA kits according to the manufacturers’ instructions. ELISA kits for KC, IL-1β, and MIP-1β were purchased from R&D Systems; ELISA kits for TNF-α and IL-1α were purchased from eBioscience; and ELISA kits for MIP-1α were purchased from Abcam.

Immunophenotyping of Ear Cells.

Ears from each mouse were dissected, disrupted mechanically using scissors, and placed in 1 mL of PBS solution containing 5% (vol/vol) FBS (Thermo Fisher Scientific) and 1 mg/mL collagenase type IV (Sigma-Aldrich). After 30 min of incubation at 37 °C, cells were centrifuged at 400 × g for 10 min, collected, and washed twice with PBS before flow cytometric analysis. Staining was performed with the fluorochrome-conjugated anti-CD11b Alexa Fluor 700 (clone M1/70), anti–Ly-6G and anti–Ly-6C (Gr1) FITC (clone RB6-8C5) monoclonal antibodies, or the corresponding isotype controls for 20 min at 4 °C in Staining Buffer (all BD Biosciences). Fluorescence was measured using a FACSAria III flow cytometer system, and the data were analyzed using FACSDiva software (all BD Biosciences).

Histological Evaluations.

Skin samples and ears were fixed in modified Davidson’s fluid and processed for histological analysis. Tissue blocks were embedded in paraffin. Series of 4-µm sections were cut through each 100 µm of tissue, and the sections were then stained with Caracci’s hematoxylin and eosin. All sections were evaluated by two pathologists blinded to the experiment’s design. For each animal, slides with the maximum inflammatory response score were selected. Microscopic images were obtained using a Keyence microscope (Model BZ-9000).

Cholesterol Sulfate Quantification in Skin.

Skin samples containing sites of DNFB or vehicle application as well as intact skin were removed using an 8-mm biopsy punch and weighed. Next the skin samples were immediately placed into tubes containing lysing matrix A (MP Biomedicals) and 0.75 mL of extraction mixture (chloroform/methanol/water, 1/2/0.8, by volume). The homogenized samples were prepared using a FastPrep 24 device and tubes containing lysing matrix A (MP Biomedicals). The homogenates were centrifuged twice at 12,000 × g for 10 min, and the supernatants were used for cholesterol sulfate quantification.

HPLC separations and cholesterol sulfate detection were conducted as described previously in the literature (49) with several important modifications. Briefly, HPLC was performed using a reversed-phase Zorbax Eclipse XBD C18, 2.1 × 150 mm, 5 µm (Agilent). The separations were performed using a Waters BioAlliance 2796 HPLC module (Waters) at room temperature with a gradient of mobile phase A [100 µM ammonium formate in 50% (vol/vol) methanol with 0.1% formic acid] and mobile phase B [100% (vol/vol) methanol]. The flow rate was 200 μL/min using gradient elution: 0–2 min, 60% B; 2–4 min, 60–90% B; 4–20 min, 90% B. The injection volume was 10 µL. The ESI-MS analyses were performed using a QSTAR Elite system (AB Sciex) equipped with a TurboIonSpray ion source. Full-scan profiling data were acquired in the TOF mass analyzer in negative ion mode, and we used the following source and ion transfer parameters: ion source nebulizer gas (GS1), 10 l/min; ion source heater gas (GS2), 5 L/min; TEM, 425 °C; and CUR, 20 L/min. The IS was −4,500 V. The DP was −70 V, and the FP was −260 V. The mass spectrometer was operated using Analyst software version 2.0 (AB Sciex). The quantitation analysis was performed in “precusor ion scan mode” in negative ion mode. In this scan type, quadrupole Q1 was scanned in the mass range of 460–470 Thomson (Th), and the ions were fragmented in the Q2 collusion cell. The ions produced were pulsed into the TOF and measured at the detector over a mass range of 50–600 Th. We used diagnostic fragment ions of 96.9700 Da and 79.9700 Da (center of precursor masses) and a mass window of 2 Da.

As a standard for quantification, we used cholesterol 3-sulfate (700016P, Avanti Polar Lipids).

Statistical Analysis.

All experiments were repeated at least twice (except for the microarray analysis). Unless stated otherwise, the statistical significance of the differences among the means was determined using a one-way ANOVA with the Statistica 7 statistical software package (StatSoft, Inc.). Differences were considered significant if P was less than 0.05.

Acknowledgments

We thank Dr. B. V. Novikov for helpful discussions and L. G. Vetkova for technical assistance with animals and the Center for Molecular and Cell Technologies, Research Park, St. Petersburg State University, and the Core Centrum of the Institute of Developmental Biology of Russian Academy of Sciences for providing technical assistance. This study was supported in part by a President’s grant from the Ministry of Education and Science of Russia (MK-5205.2015.7 to A.V.K.).

Footnotes

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

Data deposition: The microarray CEL files have been deposited in the Gene Expression Omnibus (GEO) database (accession no. GSE76144).

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1611665114/-/DCSupplemental.

References

  • 1.Bianchi ME. DAMPs, PAMPs and alarmins: All we need to know about danger. J Leukoc Biol. 2007;81(1):1–5. doi: 10.1189/jlb.0306164. [DOI] [PubMed] [Google Scholar]
  • 2.Vénéreau E, Ceriotti C, Bianchi ME. DAMPs from cell death to new life. Front Immunol. 2015;6:422. doi: 10.3389/fimmu.2015.00422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Chen GY, Nuñez G. Sterile inflammation: Sensing and reacting to damage. Nat Rev Immunol. 2010;10(12):826–837. doi: 10.1038/nri2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Józefowski S. The danger model: Questioning an unconvincing theory. Immunol Cell Biol. 2016;94(2):164–168. doi: 10.1038/icb.2015.68. [DOI] [PubMed] [Google Scholar]
  • 5.Karin M, Clevers H. Reparative inflammation takes charge of tissue regeneration. Nature. 2016;529(7586):307–315. doi: 10.1038/nature17039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Richardson MB, Williams SJ. MCL and Mincle: C-type lectin receptors that sense damaged self and pathogen-associated molecular patterns. Front Immunol. 2014;5:288. doi: 10.3389/fimmu.2014.00288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Yamasaki S, et al. Mincle is an ITAM-coupled activating receptor that senses damaged cells. Nat Immunol. 2008;9(10):1179–1188. doi: 10.1038/ni.1651. [DOI] [PubMed] [Google Scholar]
  • 8.Ostrop J, et al. Contribution of MINCLE–SYK signaling to activation of primary human APCs by mycobacterial cord factor and the novel adjuvant TDB. J Immunol. 2015;195(5):2417–2428. doi: 10.4049/jimmunol.1500102. [DOI] [PubMed] [Google Scholar]
  • 9.Schoenen H, et al. Cutting edge: Mincle is essential for recognition and adjuvanticity of the mycobacterial cord factor and its synthetic analog trehalose-dibehenate. J Immunol. 2010;184(6):2756–2760. doi: 10.4049/jimmunol.0904013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lampe MA, Williams ML, Elias PM. Human epidermal lipids: Characterization and modulations during differentiation. J Lipid Res. 1983;24(2):131–140. [PubMed] [Google Scholar]
  • 11.Rearick JI, Hesterberg TW, Jetten AM. Human bronchial epithelial cells synthesize cholesterol sulfate during squamous differentiation in vitro. J Cell Physiol. 1987;133(3):573–578. doi: 10.1002/jcp.1041330320. [DOI] [PubMed] [Google Scholar]
  • 12.Rearick JI, Stoner GD, George MA, Jetten AM. Cholesterol sulfate accumulation in tumorigenic and nontumorigenic rat esophageal epithelial cells: Evidence for defective differentiation control in tumorigenic cells. Cancer Res. 1988;48(18):5289–5295. [PubMed] [Google Scholar]
  • 13.Miyake Y, et al. C-type lectin MCL is an FcRγ-coupled receptor that mediates the adjuvanticity of mycobacterial cord factor. Immunity. 2013;38(5):1050–1062. doi: 10.1016/j.immuni.2013.03.010. [DOI] [PubMed] [Google Scholar]
  • 14.deRiveroVaccari JC, et al. Mincle signaling in the innate immune response after traumatic brain injury. J Neurotrauma. 2015;32(4):228–36. doi: 10.1089/neu.2014.3436. [DOI] [PubMed] [Google Scholar]
  • 15.He Y, et al. Macrophage-inducible C-type lectin/spleen tyrosine kinase signaling pathway contributes to neuroinflammation after subarachnoid hemorrhage in rats. Stroke. 2015;46(8):2277–2286. doi: 10.1161/STROKEAHA.115.010088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Duewell P, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010;464(7293):1357–1361. doi: 10.1038/nature08938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Tall AR, Yvan-Charvet L. Cholesterol, inflammation and innate immunity. Nat Rev Immunol. 2015;15(2):104–116. doi: 10.1038/nri3793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Röse L, Schneider C, Stock C, Zollner TM, Döcke WD. Extended DNFB-induced contact hypersensitivity models display characteristics of chronic inflammatory dermatoses. Exp Dermatol. 2012;21(1):25–31. doi: 10.1111/j.1600-0625.2011.01395.x. [DOI] [PubMed] [Google Scholar]
  • 19.Kono H, Onda A, Yanagida T. Molecular determinants of sterile inflammation. Curr Opin Immunol. 2014;26:147–156. doi: 10.1016/j.coi.2013.12.004. [DOI] [PubMed] [Google Scholar]
  • 20.Newton K, Dixit VM. Signaling in innate immunity and inflammation. Cold Spring Harb Perspect Biol. 2012;4(3):a006049. doi: 10.1101/cshperspect.a006049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Janeway CA., Jr Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb Symp Quant Biol. 1989;54(Pt 1):1–13. doi: 10.1101/sqb.1989.054.01.003. [DOI] [PubMed] [Google Scholar]
  • 22.Medzhitov R, Janeway C., Jr Innate immune recognition: Mechanisms and pathways. Immunol Rev. 2000;173:89–97. doi: 10.1034/j.1600-065x.2000.917309.x. [DOI] [PubMed] [Google Scholar]
  • 23.Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol. 1994;12:991–1045. doi: 10.1146/annurev.iy.12.040194.005015. [DOI] [PubMed] [Google Scholar]
  • 24.Matzinger P. The danger model: A renewed sense of self. Science. 2002;296(5566):301–305. doi: 10.1126/science.1071059. [DOI] [PubMed] [Google Scholar]
  • 25.Palm NW, Rosenstein RK, Medzhitov R. Allergic host defences. Nature. 2012;484(7395):465–472. doi: 10.1038/nature11047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kiyotake R, et al. Human Mincle binds to cholesterol crystals and triggers innate immune responses. J Biol Chem. 2015;290(42):25322–25332. doi: 10.1074/jbc.M115.645234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zhao XQ, et al. C-type lectin receptor dectin-3 mediates trehalose 6,6′-dimycolate (TDM)-induced Mincle expression through CARD9/Bcl10/MALT1-dependent nuclear factor (NF)-κB activation. J Biol Chem. 2014;289(43):30052–30062. doi: 10.1074/jbc.M114.588574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Tanaka M, et al. Macrophage-inducible C-type lectin underlies obesity-induced adipose tissue fibrosis. Nat Commun. 2014;5:4982. doi: 10.1038/ncomms5982. [DOI] [PubMed] [Google Scholar]
  • 29.Lee EJ, et al. Mincle activation and the Syk/Card9 signaling axis are central to the development of autoimmune disease of the eye. J Immunol. 2016;196(7):3148–3158. doi: 10.4049/jimmunol.1502355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Seifert L, et al. The necrosome promotes pancreatic oncogenesis via CXCL1 and Mincle-induced immune suppression. Nature. 2016;532(7598):245–249. doi: 10.1038/nature17403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Martin SF, et al. Mechanisms of chemical-induced innate immunity in allergic contact dermatitis. Allergy. 2011;66(9):1152–1163. doi: 10.1111/j.1398-9995.2011.02652.x. [DOI] [PubMed] [Google Scholar]
  • 32.Watanabe H, et al. Danger signaling through the inflammasome acts as a master switch between tolerance and sensitization. J Immunol. 2008;180(9):5826–5832. doi: 10.4049/jimmunol.180.9.5826. [DOI] [PubMed] [Google Scholar]
  • 33.Grabbe S, et al. Dissection of antigenic and irritative effects of epicutaneously applied haptens in mice. Evidence that not the antigenic component but nonspecific proinflammatory effects of haptens determine the concentration-dependent elicitation of allergic contact dermatitis. J Clin Invest. 1996;98(5):1158–1164. doi: 10.1172/JCI118899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bonneville M, et al. Skin contact irritation conditions the development and severity of allergic contact dermatitis. J Invest Dermatol. 2007;127(6):1430–1435. doi: 10.1038/sj.jid.5700726. [DOI] [PubMed] [Google Scholar]
  • 35.Gregorio J, et al. Plasmacytoid dendritic cells sense skin injury and promote wound healing through type I interferons. J Exp Med. 2010;207(13):2921–2930. doi: 10.1084/jem.20101102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Yasukawa S, et al. An ITAM-Syk-CARD9 signalling axis triggers contact hypersensitivity by stimulating IL-1 production in dendritic cells. Nat Commun. 2014;5:3755. doi: 10.1038/ncomms4755. [DOI] [PubMed] [Google Scholar]
  • 37.Geijtenbeek TB, Gringhuis SI. C-type lectin receptors in the control of T helper cell differentiation. Nat Rev Immunol. 2016;16(7):433–448. doi: 10.1038/nri.2016.55. [DOI] [PubMed] [Google Scholar]
  • 38.Mitsdoerffer M, et al. Proinflammatory T helper type 17 cells are effective B-cell helpers. Proc Natl Acad Sci USA. 2010;107(32):14292–14297. doi: 10.1073/pnas.1009234107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Peiser M. Role of Th17 cells in skin inflammation of allergic contact dermatitis. Clin Dev Immunol. 2013;2013:261037. doi: 10.1155/2013/261037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Deng Y, Chang C, Lu Q. The inflammatory response in psoriasis: A comprehensive review. Clin Rev Allergy Immunol. 2016;50(3):377–389. doi: 10.1007/s12016-016-8535-x. [DOI] [PubMed] [Google Scholar]
  • 41.Maddur MS, Miossec P, Kaveri SV, Bayry J. Th17 cells: Biology, pathogenesis of autoimmune and inflammatory diseases, and therapeutic strategies. Am J Pathol. 2012;181(1):8–18. doi: 10.1016/j.ajpath.2012.03.044. [DOI] [PubMed] [Google Scholar]
  • 42.Strott CA, Higashi Y. Cholesterol sulfate in human physiology: What’s it all about? J Lipid Res. 2003;44(7):1268–1278. doi: 10.1194/jlr.R300005-JLR200. [DOI] [PubMed] [Google Scholar]
  • 43.Sakharov DA, et al. Passing the anaerobic threshold is associated with substantial changes in the gene expression profile in white blood cells. Eur J Appl Physiol. 2012;112(3):963–972. doi: 10.1007/s00421-011-2048-3. [DOI] [PubMed] [Google Scholar]
  • 44.Oliveira-Ferrer L, et al. c-FOS suppresses ovarian cancer progression by changing adhesion. Br J Cancer. 2014;110(3):753–763. doi: 10.1038/bjc.2013.774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Dreyfus H, Guérold B, Freysz L, Hicks D. Successive isolation and separation of the major lipid fractions including gangliosides from single biological samples. Anal Biochem. 1997;249(1):67–78. doi: 10.1006/abio.1997.2143. [DOI] [PubMed] [Google Scholar]
  • 46.Williams MA, McCluer RH. The use of Sep-Pak C18 cartridges during the isolation of gangliosides. J Neurochem. 1980;35(1):266–269. doi: 10.1111/j.1471-4159.1980.tb12515.x. [DOI] [PubMed] [Google Scholar]
  • 47.Ikeda K, Shimizu T, Taguchi R. Targeted analysis of ganglioside and sulfatide molecular species by LC/ESI-MS/MS with theoretically expanded multiple reaction monitoring. J Lipid Res. 2008;49(12):2678–2689. doi: 10.1194/jlr.D800038-JLR200. [DOI] [PubMed] [Google Scholar]
  • 48.Inaba K, et al. Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor. J Exp Med. 1992;176(6):1693–1702. doi: 10.1084/jem.176.6.1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Fong BMW, Tam S, Leung KSY. Determination of plasma cholesterol sulfate by LC-APCI-MS/MS in the context of pediatric autism. Talanta. 2013;116:115–121. doi: 10.1016/j.talanta.2013.04.075. [DOI] [PubMed] [Google Scholar]

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES