Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Silymarin suppresses basal and stimulus-induced activation, exhaustion, differentiation, and inflammatory markers in primary human immune cells

  • Erica S. Lovelace,

    Affiliation Department of Laboratory Medicine, University of Washington, Seattle, WA, United States of America

  • Nicholas J. Maurice,

    Affiliation Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America

  • Hannah W. Miller,

    Affiliation Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America

  • Chloe K. Slichter,

    Affiliations Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America, Department of Global Health, University of Washington, Seattle, WA, United States of America

  • Robert Harrington,

    Affiliation Division of Allergy and Infectious Disease, University of Washington, Seattle, WA, United States of America

  • Amalia Magaret,

    Affiliations Department of Laboratory Medicine, University of Washington, Seattle, WA, United States of America, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America, Department of Biostatistics, University of Washington, Seattle, WA, United States of America

  • Martin Prlic,

    Affiliations Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America, Department of Global Health, University of Washington, Seattle, WA, United States of America

  • Stephen De Rosa,

    Affiliation Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America

  • Stephen J. Polyak

    polyak@uw.edu

    Affiliations Department of Laboratory Medicine, University of Washington, Seattle, WA, United States of America, Department of Global Health, University of Washington, Seattle, WA, United States of America, Department of Microbiology, University of Washington, Seattle, WA, United States of America

Abstract

Silymarin (SM), and its flavonolignan components, alter cellular metabolism and inhibit inflammatory status in human liver and T cell lines. In this study, we hypothesized that SM suppresses both acute and chronic immune activation (CIA), including in the context of HIV infection. SM treatment suppressed the expression of T cell activation and exhaustion markers on CD4+ and CD8+ T cells from chronically-infected, HIV-positive subjects. SM also showed a trend towards modifying CD4+ T cell memory subsets from HIV+ subjects. In the HIV-negative setting, SM treatment showed trends towards suppressing pro-inflammatory cytokines from non-activated and pathogen-associated molecular pattern (PAMP)-activated primary human monocytes, and non-activated and cytokine- and T cell receptor (TCR)-activated mucosal-associated invariant T (MAIT) cells. The data suggest that SM elicits broad anti-inflammatory and immunoregulatory activity in primary human immune cells. By using novel compounds to alter cellular inflammatory status, it may be possible to regulate inflammation in both non-disease and disease states.

Introduction

Inflammation is a protective and reparative response that is induced by pathogen or host-derived engagement of pattern recognition receptors (PRR) as well as through the engagement of cytokine and non-cytokine cellular receptors [1, 2]. Receptor activation triggers cellular signal transduction, causing production and release of pro-inflammatory cytokines and chemokines from cells, which in turn, recruits immune effector cells to the site of inflammation. Upon resolution of infection and/or damage, inflammatory responses normally return to baseline. Human immune cells are on the front line of many inflammatory responses, and include CD4+ and CD8+ T cells, monocytes, and mucosal associated invariant T (MAIT) cells. Temporally, monocytes and MAIT cells comprise the initial innate phase of an inflammatory response, while CD4+ and CD8+ comprise the adaptive phase and require proper inflammatory cues (from MAIT cells or monocytes) for their effector function, the quality of the immune response, and formation of a memory population. Dysregulated inflammation interrupts this regimented, temporal process.

In the case of persistent infections, dysregulated inflammation is maintained, establishing a state of chronic immune activation (CIA), which can lead to various disease states. Chronic HIV infection, despite the effective control of viremia with antiretroviral therapy (ART), is a state of CIA that leads to a host of inflammatory disorders in many infected patients [3]. With CIA, memory T cell effector functions are lost, inhibitory factors are induced, and immune cell metabolism is altered [4]. In both ART-treated and untreated HIV-infected individuals, CIA is associated with significantly elevated immune activation markers [5], various inflammatory diseases [6], cardiovascular diseases [7], both AIDS-defining and non-AIDS defining cancers [8], as well as HIV disease progression and mortality [9].

CIA in the context of HIV infection may be due to several factors [10] and can be assessed by measuring exhaustion or proliferation markers on immune cells [11], changes in immune cell inflammatory function [12], and the loss of the CD4+ T-cell population causing in an inverted CD4+/CD8+ ratio [13]. For example, the activation marker, CD38, on CD8+ and CD4+ T cells, is considered one of the best correlates for disease progression [14]. Programmed cell death protein (PD-1), which is highly expressed on exhausted T cells, is also upregulated in T cells in HIV-infected persons [15]. As such, various approaches have been used to reduce CIA including direct blockade of cellular exhaustion markers, such as targeting PD-1 and cytotoxic T lymphocyte antigen 4 (CTLA4) [16]. In addition to targeting exhaustion markers, dysregulated inflammation has also been shown to be suppressed with anti-inflammatory drugs such as aspirin [17], chloroquine [18], prednisone [6], and statins [19], all of which have been shown to reduce some parameters of CIA.

Silymarin (SM) is an herbal extract derived from the seeds of the milk thistle plant Silybum marianum [L.] Gaertn. [Asteraceae] and is frequently consumed by HCV- and HIV-infected subjects [20]. SM is known to suppress HCV infection in vitro [2125] while an intravenous formulation of silibinin (a major component of SM) inhibits HCV replication in vivo [2629], and inhibits HIV-1 infection in vitro [30]. In addition to its antiviral activities, SM suppresses various inflammation pathways: including inhibition of pro-inflammatory signaling pathways (e.g., NF-κB and forkhead box O [FOXO]), and the expression of pro-inflammatory cytokines and chemokines (e.g., CXCL1, CXCL2, CXCL8, CXCL10, IL-1, TNF-α [21, 22, 31, 32]. Furthermore, SM treatment blocks T cell activation [21, 22, 24, 33] and PHA-induced activation of peripheral blood mononuclear cells (PBMC) in vitro [30].

In this study, we explored the in vitro anti-inflammatory and immunomodulatory activities of SM in different primary human immune cells and contexts, including monocytes, MAIT cells, and T cells from HIV-infected and non-infected subjects.

Materials and methods

Silymarin preparation

Powdered extract (Product No. 345066, Lot No. 286061) of the seeds (achenes) of Silybum marianum [L.] Gaertn. was obtained from Euromed, S.A. (Barcelona, Spain), which is a part of the Madaus Group (Cologne, Germany). To eliminate stability concerns with freeze-thawing solutions of SM and the hygroscopic nature of DMSO, single use aliquots of SM were prepared as described [34, 35]. SM was reconstituted to a concentration of 10 mM in MeOH (based on a molecular weight of 482 g/mol for the seven main flavonolignan diastereoisomers). Then, 100 μL of this solution was dispensed into 0.7 mL microcentrifuge tubes and allowed to freeze-dry overnight, imparting 0.482 mg of SM per tube. The dried aliquots were stored at -20°C. Single-use aliquots of silymarin were reconstituted in 40 μL of DMSO and extensively vortexed to generate a 25 mM stock solution. DMSO solvent controls ([DMSO] ≤ 0.3%) were used for all experiments.

Monocyte and MAIT cell isolation and culture

Human monocytes were isolated from cryopreserved PBMCs from HIV-negative subjects using MACS CD14 Microbeads (Miltenyi Biotec, Bergisch Gladbach, Germany). CD14+ monocytes were rested or stimulated with LPS, a TLR4 agonist, or ssRNA, a TLR8 agonist, and then treated with SM or DMSO for 24 hours, followed by supernatant collection, and then analyzed by Luminex. Following the CD14 microbead isolation of monocytes, the remaining cells were stained with antibodies against CD161 and Va7.2 and MAIT cells were isolated by FACS [29]. MAIT cells were rested, stimulated with IL-12/15/18 (100ng/ml each) or stimulated with anti-CD3/CD28 beads (Dynabeads, Invitrogen) + IL-12/15/18 as described [36]. Rested and stimulated MAIT cells were incubated with or without SM. Cellular phenotypes were assessed based on marker expression (measured by flow cytometry) and cellular function was based on cytokine production (measured by Intracellular Cytokine Stain [ICS] assay).

PBMC samples and culturing

PBMC samples from HIV-infected subjects were obtained from the University of Washington (UW) Center for AIDS Research (CFAR) Specimen Repository. All PBMC specimens are linked to comprehensive clinical data within the UW HIV Information System allowing the identification of patients who HIV-positive and receiving ART, or HIV-positive and ART-naïve or not on ART at the time of donation. Inclusion and exclusion criteria and subject characteristics are listed in Tables 1 and 2.

thumbnail
Table 1. Inclusion and Exclusion Criteria for selecting PBMC samples.

https://doi.org/10.1371/journal.pone.0171139.t001

thumbnail
Table 2. Characteristics of Subjects Whose PBMCs Were Analyzed by Flow Cytometry.

https://doi.org/10.1371/journal.pone.0171139.t002

HIV+ cryopreserved PBMC were thawed in a 37°C water bath, transferred to a 50 ml conical tube containing 10 ml of R10 medium (RPMI 1640 with 25 mM HEPES buffer and L-glutamate [Gibco BRL Life Technologies, Waltham, MA, USA; 10% fetal bovine serum [FBS; Gemini Bio-products, West Sacramento, CA, USA], and Penicillin-Streptomcyin [Gibco BRL Life Technologies, Waltham, MA, USA]), centrifuged at 250 x g for 10 minutes, and the cell pellet was resuspended in R10 medium. Cell count and viability were determined using a Guava Viacount (EMD Millipore, Billerica, MA, USA), resuspended to approximately 2 x 106 cells/ml in PBS (Gibco BRL Life Technologies, Waltham, MA, USA), and stained for flow cytometry analysis ex vivo or prepared for culture including treatment with SM (at 80 μM) or DMSO (as a vehicle control) for 72 hours at 37°C. After 72 hours in culture, cell counts and viability were again measured prior to marker staining and flow cytometry. Thirty-three samples were initially evaluated, and eight samples were excluded because of low viability. Thus, statistical analyses are based on samples from 25 subjects.

Eleven-color polychromatic flow cytometry

Using an 11-marker flow cytometry antibody panel (listed in Table 3), we assessed CD4+ and CD8+ T cell phenotypes by measuring the expression of activation and exhaustion markers in HIV+ PBMC. We adopted our protocol as described previously [37]. Samples were stained with Aqua Live/Dead Fixable Dead Cell Stain (AViD; [38]) then surface-stained with antibodies against CD25, CD38, CD127, CTLA4, HLA-DR, and PD1. Cells were then fixed and permeabilized using FIX & PERM Cell Fixation and Cell Permeabilization Kit (ThermoFisher Scientific, Waltham, MA, USA). Cells were stored at -80°C in the dark overnight in FACS Wash (PBS with 2% FBS) following permeabilization. The following day, cells were thawed at 37°C for 10 minutes, and then intracellular staining was performed with antibody against CD3, CD4, CD8, and Ki67. Following intracellular staining, cells were stored at 4°C in PBS with 2% paraformaldehyde until samples were acquired on an LSRII BD flow cytometer using BD FACSDiva software (BD Biosciences). Flow cytometric analyses were performed using FlowJo version 9 (Tree Star). Criteria for evaluable responses were determined as previously described [39].

thumbnail
Table 3. Flow Cytometry Panel Used to Determine Activation and Exhaustion in PBMC Samples.

https://doi.org/10.1371/journal.pone.0171139.t003

S1 Fig shows the gating and staining strategy to identify CD4+ and CD8+ cells and activation and exhaustion markers. A timing gate was used to minimize exclusion events, and dead cells were excluded using the AViD (i.e., amine reactive dye) stain. In addition, single cell populations were gated, and within the lymphocyte gate, CD3+ cells were identified followed by gating on CD4+ or CD8+ cells. Using the OMIP-025 panel [48], a subset of HIV+ PBMC samples (N = 4) were separately stained using a T cell differentiation and memory panel. All HIV+ PBMC, in both panels, were stained with LIVE/DEAD® Fixable Aqua Dead Cell Stain Kit (ThermoFisher Scientific, Waltham, MA, USA), washed, and stained with the activation/exhaustion or differentiation/memory staining panels.

Luminex experiments

Seven chronically-infected, HIV+ PBMC samples were thawed and treated with SM (80 μM) or DMSO (vehicle control), cultured for 72 hours, with supernatants added to a final 1% Triton-X (Sigma-Aldrich, St. Louis, MO, USA) concentration, and then processed for Luminex analysis. The Cytokine Laboratory, a Shared Resource of the Fred Hutchinson Cancer Research Center, processed the Luminex samples.

Statistical analyses

For MAIT cells and monocyte experiments, differences in cytokine production were assessed by paired Wilcoxon signed rank tests. Twenty-five PBMC samples were included in the final flow cytometry analyses. We measured changes in marker expression on cells with SM treatment relative to DMSO alone, and computed absolute differences in percentages of marker expression (i.e., SM minus DMSO treatments). Subsequently, we assessed non-parametrically whether differences in expected expression tended to be statistically significantly different from zero, using a Wilcoxon signed rank test. Differences in expression with SM treatment minus expression with DMSO-only treatment are negative if SM reduced expression and positive if it increased expression. For markers showing a significant expression reduction with SM (versus DMSO), we also used Spearman’s correlation to assess whether expression reduction was associated with continuous measures like age and CD4:CD8 ratio; and we used Wilcoxon rank sum test to assess whether expression reduction was associated with binary factors like ART use.

Results

We have previously shown that SM prevents activation and inflammatory responses in CD4+ and CD8+ T cells from both non-infected and HCV-infected subjects [21, 22, 32]. Therefore, in the current study, we first investigated the effects of SM on other primary human immune cells from non-infected donors stimulated in the context of different triggers of inflammation. SM treatment showed a trend for reduced levels of MIP1-α, MIP1-β, IL-1α, IL1-β, RANTES, and TNF-α production from both non-stimulated and PAMP-stimulated monocytes (Fig 1). Due to the small sample size (i.e., 3 donors), the observed effect did not reach statistical significance.

thumbnail
Fig 1. SM inhibits basal and PAMP-induction of pro-inflammatory cytokines from primary human monocytes.

Luminex analysis of MIP1α (A), MIP1β (B), TNF-α (C), IL1α (D), IL1β (E), and RANTES (F) from supernatants collected from CD14+ monocytes cultured in the absence or presence of SM (80 μM) for 24 hours in different activating conditions: rested (black), LPS-stimulated, a TLR4 agonist (dark gray), or ssRNA-stimulated, a TLR8 agonist (light gray). Data shown are from three different donors, and data are displayed as mean +/- SEM. Dotted line represents limit of detection (LOD).

https://doi.org/10.1371/journal.pone.0171139.g001

Mucosal-associated invariant T (MAIT) cells have a memory-like phenotype, make up one to eight percent of circulating T cells, and are activated by IL-12, 15, and 18 to produce pro-inflammatory cytokines TNF-α and IFN-γ, and granzyme B [49, 50]. SM treatment also showed a trend for suppressing basal, cytokine (IL-12, 15, and 18), and cytokine and TCR-mediated (IL-12, 15, and 18 plus anti-CD3/28 beads) IFN-γ and Granzyme B in CD8+ MAIT cells (Fig 2). For this small sample size of three donors, the effects did not reach statistical significance. The data suggest that SM treatment suppresses inflammation in resting and PAMP-, cytokine-, and TCR-activated primary human monocyte and CD8+ MAIT cell types.

thumbnail
Fig 2. SM suppresses inflammation in resting and cytokine- and TCR-activated MAIT cells.

Expression of IFN-γ (A) and granzyme B (B) by CD8+ Va7.2+ CD161hi sorted MAIT cells cultured for 24 hours at rest, 100ng/ml IL-12, 15, and 18, or a combination of IL-12, 15, 18 and anti-CD3/CD28 beads, in the presence or absence of SM (80 μM). Data shown are from three different donors, and data are displayed as mean +/- SEM.

https://doi.org/10.1371/journal.pone.0171139.g002

In the current study, we interrogated expression of traditional activation markers of CIA (e.g., HLA-DR and CD38), as well as other markers of T-cell activation (e.g., Ki-67 [30]) and markers for T-cell exhaustion, including PD-1 and CTLA-4 [40] using our flow cytometry panel (Table 3). We analyzed the effect of SM treatment on T cell marker expression from 25 human PBMC samples (Table 2) including: four HIV-negative subjects, and 21 subjects who were HIV-positive (i.e., 14 subjects were on ART and seven subjects were not on ART). We used a dose of 80μM SM, which we have previously shown to be non-toxic to human cells [31]. We monitored cell viability of all PBMC cultures, determined as the percentage of AViD-negative cells. Using a Wilcoxon paired test, the 80μM dose of SM, after 72 hours of ex vivo cell culture, induced a small, but significant (p<0.0001) decrease in viability: the median decrease of viability due to SM treatment relative to DMSO was 7%, (IQR [2%–12%]). Twenty-five PBMC samples had greater than 50% viability and were included in statistical analyses. Moreover, all flow cytometry data described below are based on viable cells.

SM treatment reduced the expression of activation markers in HIV-negative and HIV-positive T cells (Table 4 and S2 Fig). Specifically, SM treatment significantly reduced the expression of CD38+/HLA-DR+ (compared to DMSO) in both CD4+ and CD8+ T cells (i.e., difference of -0.7%; median IQR [-1.1% to -0.4%]; p-value = 0.001) and (-0.6%; [-1.7% to -0.3%]; p-value = 0.0003), respectively (Fig 3). SM caused a greater suppression of CD38+/HLA-DR+ expression on T cells from ART-naïve patients as compared to patients on ART. This effect was observed on both CD4+ (p-value = 0.001) and CD8+ (p-value = 0.0074) T cells (S3A–S3D Fig).

thumbnail
Fig 3. SM suppresses the expression of activation and exhaustion markers on CD4+ and CD8+ T cells from HIV+ subjects.

Expression of activation markers, CD38 and HLA-DR on CD4+ (A) and CD8+ (B) T cells, and exhaustion markers, CTLA4 and PD1, on CD4+ (C) and CD8+ (D) T cells from PBMC cultures treated for 72 hours with SM (80 μM; empty symbols) or DMSO (vehicle control; solid symbols). Data are from 25 different PBMC samples. P values are derived from Wilcoxon signed rank tests.

https://doi.org/10.1371/journal.pone.0171139.g003

thumbnail
Table 4. SM Suppresses T Cell Activation and Exhaustion Markers.

https://doi.org/10.1371/journal.pone.0171139.t004

In the CD4+ T cell population, the median difference in CD38+/HLA-DR+ expression between SM and DMSO treated cells was -1.3% (IQR -1.8% to -1.1%) for ART-naïve individuals and was -0.5% (IQR -0.7% to -0.2%) for subjects on ART (S2A and S2B Fig). In the CD8+ T cell population, the median difference of CD38+ and HLA-DR+ expression between SM and DMSO treated cells was -3.3% (-8.0% to -0.9%) for ART-naïve individuals and was 0.4% (IQR -0.8% to 0.1%) for subjects on ART (S2C and S2D Fig). Lower CD4:CD8 ratio was associated with a significant decrease in CD38+ and HLA-DR+ expression on SM versus DMSO treated CD8+ T cells (Spearman’s rho = 0.65, p-value = 0.002; S2E and S2F Fig) but was not significantly associated with a decrease on CD4+ T cells (Spearman’s rho = 0.37, p = 0.095). There were no age-related associations in the difference of CD38+/HLA-DR+ expression for either CD4+ or CD8+ T cells (p = 0.27 and p = 0.34, respectively).

SM treatment also suppressed the expression of T cell exhaustion markers, CTLA4 and PD1 (Table 4 and S3 Fig). Specifically, CTLA4+/PD1+ expression on CD4+ T cells was significantly reduced by SM treatment in comparison to DMSO (difference of -0.9%; IQR [21.9% to 0.5%]; p-value = 0.037; Table 4, Fig 3). These effects were limited to CD4+ T cells, as SM did not significantly reduce these markers on CD8+ T cells (p = 0.32). Moreover, the reduction of CTLA4+/PD1+ on CD4+ T cells by SM treatment was strongly associated with CD4:CD8 ratio (Spearman’s rho = 0.57; p-value = 0.010; S3A and S3B Fig). As with CD38+/HLA-DR+ markers, no association of CTLA4+/PD1+ markers was observed with age (p = 0.79). However, SM reduction of CTLA4+/PD1+ markers on CD4+ T cells was greater in magnitude in PBMC samples from individuals that were ART-naïve (median difference of -24.2%; IQR [-31.8–21.0%]) versus those that were receiving ART (-0.1% [IQR -6.4% to 0.4%], Wilcoxon test, p = 0.037) (S3C and S3D Fig).

After 72 hours in culture, SM did not significantly alter the expression of the activation marker Ki67 in CD4+ and CD8+ T cells (Table 4; S4A and S4B Fig). In addition, SM treatment did not have any significant effect on CD4+ regulatory T cell markers (i.e., Tregs; defined as CD25+/CD127lo CD4+ T cells; p-value = 0.71), while SM treatment marginally reduced the expression of CD25+/CD127lo on CD8+ T cells (p-value = 0.046) (Table 4; S4C and S4D Fig).

To determine if SM treatment results in the inhibition of T cell pro-inflammatory function, a subset of chronically infected, HIV+ PBMC culture supernatants were harvested after 72 hours of culture in SM or DMSO and processed for Luminex analyses. As shown in S5 Fig, SM treatment suppressed IL-18 and IL-6 expression in cultures, whereas the effect of SM on IL-8 expression was variable.

The chemokine receptor CCR7 and the tyrosine phosphatase CD45RA are used to designate T cells as naïve (CD45RA+/CCR7+), terminal effector (CD45RA+/CCR7-), central memory (CD45RA-/CCR7+), and effector memory (CD45RA-/CCR7-) cells [51, 52]. While not statistically significant due to the low number of samples (N = 4), SM treatment showed a trend for increasing the percentage of naïve CD4+ T cells (defined as CD4+ cells expressing CD45RA+/CCR7+ markers) in comparison to DMSO treatment (Fig 4). SM treatment also showed a trend towards reducing the percentage of CD4 T cells in the terminal effector and effector memory cell subsets.

thumbnail
Fig 4. SM affects the differentiation states of CD4+ T cell populations in HIV+ PBMC cultures.

After 72 hours of treatment with SM (80 μM; empty symbols) or DMSO (vehicle control; solid symbols), PBMC were analyzed by flow cytometry for CD4+ T cell differentiation markers, defined as follows: A, Naïve (CD45RA+/CCR7+), B, Terminal Effector (CD45RA+/CCR7-), C, Effector Memory (CD45RA-/CCR7-), and D, Central Memory (CD45RA-/CCR7+).

https://doi.org/10.1371/journal.pone.0171139.g004

Discussion

In this study, we show that SM significantly reduces expression of various markers of T cell activation and exhaustion (e.g., CD38, HLA-DR, CTLA4, and PD1) as well as pro-inflammatory cytokines (e.g., IL-6 and IL-18) in PBMCs from chronically-infected HIV-positive subjects. SM treatment also showed a trend towards maintaining naïve CD4+ T cell populations in cultured PBMC from chronically infected, HIV+ subjects. For monocytes and MAIT cells in the HIV-negative setting, SM treatment showed trends toward reducing pro-inflammatory cytokines in resting/non-stimulated, and cytokine-, PAMP-, and TCR-activated cultures. While interesting, these effects of SM on monocytes, MAIT cells, and T cell memory subsets need to be bolstered in future studies by studying larger numbers of samples. Nevertheless, the data indicate that the inflammatory status of primary human immune cells, in both resting and stimulus-activated conditions, is suppressed by SM.

Similar to activation and exhaustion markers, SM suppression of PAMP-induced inflammation in monocytes may be important during CIA. For example, monocyte activation has been associated with systemic inflammation, atherosclerosis, and the development of premature cardiovascular disease [53], all of which occur at a higher rate in the HIV-infected community [54, 55].

The effects of SM treatment may not only be applicable to monocytes and T cells in the context of HIV infection, as SM treatment may also be effective for the suppression of MAIT activation involved in resetting pathologic inflammation. Multiple studies have shown that CD8+ MAITs contribute to the primary, innate-like response against bacterial and fungal infections and can be activated by inflammatory stimuli [36, 56]. The suppression of CD8+ MAIT cell activation, as shown here by SM treatment, is not limited to that caused by acute bacterial infections, but may also be important during chronic viral infections, such as HIV: circulating MAIT cells are severely reduced in chronically-infected HIV+ patients, and fail to completely restore to pre-infection frequencies with ART [57]. Moreover, these MAIT cells showed a functionally exhausted phenotype. Our data suggest that by treating MAIT cells with SM and reducing their inflammatory response, it may be possible to reduce the MAIT-associated exhausted phenotype, preserving their function in both the blood and at epithelial sites, such as in the context of chronic viral infections.

The reduction of the T cell activation markers CD38 and HLA-DR by SM was correlated with a low CD4:CD8 ratio (i.e., < 1) and in PBMC from subjects not on ART. In the setting of chronic HIV infection, clinical parameters such as CD4:CD8 ratio have been used to identify HIV+ individuals on ART who are at greater risk of immune dysfunction, leading to AIDS, non-AIDS events, and mortality [58]. Thus, natural products like SM may be effective in reducing CIA that is associated with AIDS, non-AIDS events, and mortality. In addition to the traditional activation markers CD38+/HLA-DR+, the reduction of exhaustion markers CTLA4 and PD1 by SM may also be clinically relevant in the context of HIV-associated CIA. For example, blocking PD-1 receptor function during simian immunodeficiency virus in vivo was shown to improve virus-specific CD8+ T cell responses, reduce plasma viremia, as well as increase animal survival [59]. In fact, by inhibiting multiple immune exhaustion pathways (e.g., PD-1 and CTLA4) simultaneously, T cell function can further be improved [60], suggesting potential additive or synergistic therapies for recovery of immune function. In the context of chronic HIV infection, the ability of SM to reduce CTLA4 and PD1 expression is particularly interesting, since cells with high-level expression of exhaustion markers may preferentially harbor latent, HIV proviral DNA [61].

The ability of SM to quell various types of inflammation is similar to the immunomodulatory effects of metabolic modulators like rapamycin, which can suppress cellular activation, stimulus-induced inflammation, and alter immune cell fate (i.e. differentiation). Rapamycin’s principle mechanism of action is via inhibition of mammalian target of rapamycin (mTOR) through its association with FKBP12 [62, 63]. We have recently shown that SM also inhibits mTOR by at least two pathways, involving activation of adenosine monophosphate kinase (AMPK) and induction of DNA Damage Inducible Transcript 4 (DDIT4), a novel inhibitor of mTOR [31]. Moreover, SM activates AMPK signaling, which feeds forward to suppress NF-κB signaling [31]. Thus, SM appears to modulate multiple metabolic pathways that converge on suppressing inflammation in diverse immune cell types, independent of the inflammatory insult or stimulus.

Supporting information

S1 Fig. Example of gating tree used for flow cytometry data.

A representative example of a PBMC sample, cultured with DMSO (vehicle control) for 72 hours, stained with the immune exhaustion panel. Gating is as follows: top row, Time, Singlet, AViD Live/Dead. Second row, Lymphocyte, CD3+, CD4+ and CD8+ Cells. Third and forth rows, CD4+ and CD8+ cells, respectively, gated for CD25hi/CD127lo, CD38+/HLA-DR+, Ki67hi, and CTLA4+/PD1+.

https://doi.org/10.1371/journal.pone.0171139.s001

(PDF)

S2 Fig. Relationship of SM Suppression of CD38+/HLA-DR+ in CD4+ and CD8+ T cells with ART status and CD4:CD8 ratio.

Panels A and B, correlations of SM suppression of activation markers CD38 and HLA-DR on CD4+ T cells with ART status. Panel A is the actual data while panel B plots the difference. Panels C and D, correlations of SM suppression of activation markers CD38 and HLA-DR on CD8+ T cells with ART status. Panel C is the actual data while panel D plots the difference. Panels E and F, association of SM suppression of CD38 and HLA-DR on CD8+ T cells with CD4:CD8 ratio.

https://doi.org/10.1371/journal.pone.0171139.s002

(PDF)

S3 Fig. Suppression of CTLA4/PD1 on CD4+ T cells is associated with CD4:CD8 ratio and ART status.

Panels A and B, difference in CTLA/PD1 expression was strongly associated with CD4:8 ratio for CD4+ T cells. Panels C and D, SM reduction of CTLA/PD1 on CD4+ T cells was greater in samples from ART-naïve individuals.

https://doi.org/10.1371/journal.pone.0171139.s003

(PDF)

S4 Fig. SM does not alter the expression of activation (Ki67) and T regulatory cell (Treg) markers on PBMC from HIV-infected individuals.

PBMC were thawed and cultured for 72 hours in either SM (80 μM; empty symbols) or DMSO (vehicle control; solid symbols) followed by staining with the exhaustion panel (listed in Table 3). Samples were assessed for the activation marker Ki67 on CD4+ (A) and CD8+ (B) T cells, and Treg markers, defined as expression of CD25+/CD127lo on CD4+ (C) and CD8+ (D) T cells.

https://doi.org/10.1371/journal.pone.0171139.s004

(PDF)

S5 Fig. SM suppresses pro-inflammatory cytokines in HIV+ PBMC cultures.

PBMC were thawed and treated with SM (80 μM) or DMSO (vehicle control), cultured for 72 hours, with supernatants lysed by the addition of a final 1% Triton-X concentration, and then processed for Luminex analysis. Data shown are from seven different PBMC samples.

https://doi.org/10.1371/journal.pone.0171139.s005

(PDF)

Acknowledgments

We would like to thank Michalina Monsanto for assistance and helpful discussions regarding PBMC repository inquiries. We thank Nicholas Oberlies and Tyler Graf for providing SM, and Rick Lawler for performing the Luminex experiments. This work was partially supported by NIH grants 5R01AT006842 from NCCIH (to S.J.P.) and T32 AI007509 (to C.K.S). Support was also provided by pilot funding from the University of Washington Center for AIDS Research under National Institutes of Health (NIH) award number P30AI027757 (from NIAID, NCI, NIMH, NIDA, NICHD, NHLBI, NIA, NIGMS, NIDDK).

Author Contributions

  1. Conceptualization: ESL NJM HWM RH AM MP SDR SJP.
  2. Formal analysis: ESL NJM HWM CKS AM MP SJP.
  3. Funding acquisition: SJP CKS.
  4. Investigation: ESL NJM HWM CKS AM.
  5. Methodology: ESL NJM HWM CKS RH AM MP SDR.
  6. Project administration: SJP.
  7. Resources: MP RH AM MP SDR SJP.
  8. Supervision: SJP.
  9. Validation: ESL NJM HWM CKS AM MP SDR SJP.
  10. Visualization: ESL AM SJP.
  11. Writing – original draft: ESL SJP AM.
  12. Writing – review & editing: ESL NJM HWM CKS RH AM MP SDR SJP.

References

  1. 1. Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nat Immunol. 2004;5(10):987–95. pmid:15454922
  2. 2. Chen GY, Nunez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol. 2010;10(12):826–37. PubMed Central PMCID: PMCPMC3114424. pmid:21088683
  3. 3. Taiwo B, Barcena L, Tressler R. Understanding and controlling chronic immune activation in the HIV-infected patients suppressed on combination antiretroviral therapy. Curr HIV/AIDS Rep. 2013;10(1):21–32. pmid:23225316
  4. 4. McKinney EF, Smith KG. T-cell exhaustion: understanding the interface of chronic viral and autoinflammatory diseases. Immunol Cell Biol. 2016;94(10):935–42. pmid:27577866
  5. 5. Hunt PW, Martin JN, Sinclair E, Bredt B, Hagos E, Lampiris H, et al. T cell activation is associated with lower CD4+ T cell gains in human immunodeficiency virus-infected patients with sustained viral suppression during antiretroviral therapy. J Infect Dis. 2003;187(10):1534–43. pmid:12721933
  6. 6. McComsey GA, Whalen CC, Mawhorter SD, Asaad R, Valdez H, Patki AH, et al. Placebo-controlled trial of prednisone in advanced HIV-1 infection. AIDS. 2001;15(3):321–7. Epub 2001/03/29. pmid:11273211
  7. 7. Nordell AD, McKenna M, Borges AH, Duprez D, Neuhaus J, Neaton JD, et al. Severity of cardiovascular disease outcomes among patients with HIV is related to markers of inflammation and coagulation. J Am Heart Assoc. 2014;3(3):e000844. PubMed Central PMCID: PMCPMC4309077. pmid:24870935
  8. 8. Ipp H, Zemlin AE, Erasmus RT, Glashoff RH. Role of inflammation in HIV-1 disease progression and prognosis. Crit Rev Clin Lab Sci. 2014;51(2):98–111. pmid:24479745
  9. 9. d'Ettorre G, Paiardini M, Ceccarelli G, Silvestri G, Vullo V. HIV-associated immune activation: from bench to bedside. AIDS Res Hum Retroviruses. 2011;27(4):355–64. Epub 2011/02/12. pmid:21309730
  10. 10. Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12(12):1365–71. Epub 2006/11/23. pmid:17115046
  11. 11. Ho HN, Hultin LE, Mitsuyasu RT, Matud JL, Hausner MA, Bockstoce D, et al. Circulating HIV-specific CD8+ cytotoxic T cells express CD38 and HLA-DR antigens. Journal of immunology. 1993;150(7):3070–9. Epub 1993/04/01.
  12. 12. Kamat A, Misra V, Cassol E, Ancuta P, Yan Z, Li C, et al. A plasma biomarker signature of immune activation in HIV patients on antiretroviral therapy. PloS one. 2012;7(2):e30881. Epub 2012/03/01. PubMed Central PMCID: PMC3281899. pmid:22363505
  13. 13. Margolick JB, Gange SJ, Detels R, O'Gorman MR, Rinaldo CR Jr., Lai S. Impact of inversion of the CD4/CD8 ratio on the natural history of HIV-1 infection. Journal of acquired immune deficiency syndromes. 2006;42(5):620–6. Epub 2006/07/27. pmid:16868499
  14. 14. Giorgi JV, Detels R. T-cell subset alterations in HIV-infected homosexual men: NIAID Multicenter AIDS cohort study. Clin Immunol Immunopathol. 1989;52(1):10–8. Epub 1989/07/01. pmid:2656013
  15. 15. Utay NS, Hunt PW. Role of immune activation in progression to AIDS. Curr Opin HIV AIDS. 2016;11(2):131–7. PubMed Central PMCID: PMCPMC4750472. pmid:26731430
  16. 16. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439(7077):682–7. pmid:16382236
  17. 17. O'Brien M, Montenont E, Hu L, Nardi MA, Valdes V, Merolla M, et al. Aspirin attenuates platelet activation and immune activation in HIV-infected subjects on antiretroviral therapy: A Pilot Study. Journal of acquired immune deficiency syndromes. 2013. Epub 2013/02/15.
  18. 18. Murray SM, Down CM, Boulware DR, Stauffer WM, Cavert WP, Schacker TW, et al. Reduction of immune activation with chloroquine therapy during chronic HIV infection. Journal of virology. 2010;84(22):12082–6. Epub 2010/09/17. PubMed Central PMCID: PMC2977889. pmid:20844049
  19. 19. De Wit S, Delforge M, Necsoi CV, Clumeck N. Downregulation of CD38 activation markers by atorvastatin in HIV patients with undetectable viral load. AIDS. 2011;25(10):1332–3. Epub 2011/04/22. pmid:21508802
  20. 20. Seeff LB, Curto TM, Szabo G, Everson GT, Bonkovsky HL, Dienstag JL, et al. Herbal product use by persons enrolled in the hepatitis C Antiviral Long-Term Treatment Against Cirrhosis (HALT-C) Trial. Hepatology. 2008;47(2):605–12. Epub 2007/12/25. pmid:18157835
  21. 21. Polyak SJ, Morishima C, Lohmann V, Pal S, Lee DY, Liu Y, et al. Identification of hepatoprotective flavonolignans from silymarin. Proc Natl Acad Sci U S A. 2010;107(13):5995–9. PubMed Central PMCID: PMC2851903. pmid:20231449
  22. 22. Polyak SJ, Morishima C, Shuhart MC, Wang CC, Liu Y, Lee DY. Inhibition of T-cell inflammatory cytokines, hepatocyte NF-kappaB signaling, and HCV infection by standardized Silymarin. Gastroenterology. 2007;132(5):1925–36. Epub 2007/05/09. pmid:17484885
  23. 23. Polyak SJ, Ferenci P, Pawlotsky JM. Hepatoprotective and antiviral functions of silymarin components in hepatitis C virus infection. Hepatology. 2013;57(3):1262–71. PubMed Central PMCID: PMC3594650. pmid:23213025
  24. 24. Wagoner J, Morishima C, Graf TN, Oberlies NH, Teissier E, Pecheur EI, et al. Differential in vitro effects of intravenous versus oral formulations of silibinin on the HCV life cycle and inflammation. PLoS One. 2011;6(1):e16464. PubMed Central PMCID: PMC3030583. pmid:21297992
  25. 25. Wagoner J, Negash A, Kane OJ, Martinez LE, Nahmias Y, Bourne N, et al. Multiple effects of silymarin on the hepatitis C virus lifecycle. Hepatology. 2010;51(6):1912–21. PubMed Central PMCID: PMC2909978. pmid:20512985
  26. 26. Beinhardt S, Rasoul-Rockenschaub S, Maieron A, P.H. S-M , Hofer H, P F. Intravenous Silibinin-therapy in patients with chronic hepatitis C in the transplant setting. J Hepatology. 2012;56(Suppl 1):S77.
  27. 27. Beinhardt S, Rasoul-Rockenschaub S, Scherzer TM, Ferenci P. Silibinin monotherapy prevents graft infection after orthotopic liver transplantation in a patient with chronic hepatitis C. Journal of hepatology. 2011;54(3):591–2; author reply 2–3. Epub 2010/11/26. pmid:21106270
  28. 28. Ferenci P, Scherzer TM, Kerschner H, Rutter K, Beinhardt S, Hofer H, et al. Silibinin is a potent antiviral agent in patients with chronic hepatitis C not responding to pegylated interferon/ribavirin therapy. Gastroenterology. 2008;135(5):1561–7. Epub 2008/09/06. pmid:18771667
  29. 29. Neumann UP, Biermer M, Eurich D, Neuhaus P, Berg T. Successful prevention of hepatitis C virus (HCV) liver graft reinfection by silibinin mono-therapy. J Hepatol. 2010;52(6):951–2. pmid:20413176
  30. 30. McClure J, Lovelace ES, Elahi S, Maurice NJ, Wagoner J, Dragavon J, et al. Silibinin Inhibits HIV-1 Infection by Reducing Cellular Activation and Proliferation. PLoS One. 2012;7(7):e41832. pmid:22848626
  31. 31. Lovelace ES, Wagoner J, MacDonald J, Bammler T, Bruckner J, Brownell J, et al. Silymarin Suppresses Cellular Inflammation By Inducing Reparative Stress Signaling. J Nat Prod. 2015.
  32. 32. Morishima C, Shuhart MC, Wang CC, Paschal DM, Apodaca MC, Liu Y, et al. Silymarin inhibits in vitro T-cell proliferation and cytokine production in hepatitis C virus infection. Gastroenterology. 2010;138(2):671–81, 81 e1-2. PubMed Central PMCID: PMC2819600. pmid:19782083
  33. 33. Polyak SJ, Oberlies NH, Pecheur EI, Dahari H, Ferenci P, Pawlotsky JM. Silymarin for HCV infection. Antivir Ther. 2013;18(2):141–7. pmid:23011959
  34. 34. Graf TN, Cech NB, Polyak SJ, Oberlies NH. A validated UHPLC-tandem mass spectrometry method for quantitative analysis of flavonolignans in milk thistle (Silybum marianum) extracts. J Pharm Biomed Anal. 2016;126:26–33. PubMed Central PMCID: PMCPMC4893890. pmid:27136284
  35. 35. Graf TN, Wani MC, Agarwal R, Kroll DJ, Oberlies NH. Gram-scale purification of flavonolignan diastereoisomers from Silybum marianum (Milk Thistle) extract in support of preclinical in vivo studies for prostate cancer chemoprevention. Planta Med. 2007;73(14):1495–501. pmid:17948171
  36. 36. Slichter CK, McDavid A, Miller HW, Finak G, Seymour BJ, McNevin JP, et al. Distinct activation thresholds of human conventional and innate-like memory T cells. JCI Insight. 2016;1(8). PubMed Central PMCID: PMCPMC4912124.
  37. 37. De Rosa SC, Thomas EP, Bui J, Huang Y, deCamp A, Morgan C, et al. HIV-DNA priming alters T cell responses to HIV-adenovirus vaccine even when responses to DNA are undetectable. Journal of immunology. 2011;187(6):3391–401. Epub 2011/08/17. PubMed Central PMCID: PMC3180898.
  38. 38. Perfetto SP, Chattopadhyay PK, Lamoreaux L, Nguyen R, Ambrozak D, Koup RA, et al. Amine reactive dyes: an effective tool to discriminate live and dead cells in polychromatic flow cytometry. J Immunol Methods. 2006;313(1–2):199–208. pmid:16756987
  39. 39. Horton H, Thomas EP, Stucky JA, Frank I, Moodie Z, Huang Y, et al. Optimization and validation of an 8-color intracellular cytokine staining (ICS) assay to quantify antigen-specific T cells induced by vaccination. J Immunol Methods. 2007;323(1):39–54. PubMed Central PMCID: PMCPMC2683732. pmid:17451739
  40. 40. Kanellopoulos JM, Wigglesworth NM, Owen MJ, Crumpton MJ. Biosynthesis and molecular nature of the T3 antigen of human T lymphocytes. Embo J. 1983;2(10):1807–14. PubMed Central PMCID: PMC555363. pmid:6227478
  41. 41. Dalgleish AG, Beverley PC, Clapham PR, Crawford DH, Greaves MF, Weiss RA. The CD4 (T4) antigen is an essential component of the receptor for the AIDS retrovirus. Nature. 1984;312(5996):763–7. pmid:6096719
  42. 42. Thomas Y, Rogozinski L, Rabbani L, Chess A, Goldstein G, Chess L. Functional analysis of human T cell subsets defined by monoclonal antibodies. VI. Distinct and opposing immunoregulatory functions within the OKT8+ population. J Mol Cell Immunol. 1984;1(2):103–13. pmid:6152892
  43. 43. Kinter AL, Hennessey M, Bell A, Kern S, Lin Y, Daucher M, et al. CD25(+)CD4(+) regulatory T cells from the peripheral blood of asymptomatic HIV-infected individuals regulate CD4(+) and CD8(+) HIV-specific T cell immune responses in vitro and are associated with favorable clinical markers of disease status. J Exp Med. 2004;200(3):331–43. PubMed Central PMCID: PMC2211981. pmid:15280419
  44. 44. Levacher M, Hulstaert F, Tallet S, Ullery S, Pocidalo JJ, Bach BA. The significance of activation markers on CD8 lymphocytes in human immunodeficiency syndrome: staging and prognostic value. Clin Exp Immunol. 1992;90(3):376–82. Epub 1992/12/01. PubMed Central PMCID: PMC1554578. pmid:1458674
  45. 45. Paiardini M, Cervasi B, Albrecht H, Muthukumar A, Dunham R, Gordon S, et al. Loss of CD127 expression defines an expansion of effector CD8+ T cells in HIV-infected individuals. J Immunol. 2005;174(5):2900–9. pmid:15728501
  46. 46. Kaufmann DE, Kavanagh DG, Pereyra F, Zaunders JJ, Mackey EW, Miura T, et al. Upregulation of CTLA-4 by HIV-specific CD4+ T cells correlates with disease progression and defines a reversible immune dysfunction. Nature immunology. 2007;8(11):1246–54. Epub 2007/10/02. pmid:17906628
  47. 47. Cohen Stuart JW, Hazebergh MD, Hamann D, Otto SA, Borleffs JC, Miedema F, et al. The dominant source of CD4+ and CD8+ T-cell activation in HIV infection is antigenic stimulation. Journal of acquired immune deficiency syndromes. 2000;25(3):203–11. Epub 2000/12/15. pmid:11115950
  48. 48. Moncunill G, Dobano C, McElrath MJ, De Rosa SC. OMIP-025: evaluation of human T- and NK-cell responses including memory and follicular helper phenotype by intracellular cytokine staining. Cytometry A. 2015;87(4):289–92. PubMed Central PMCID: PMCPMC4454451. pmid:25407958
  49. 49. Gapin L. Check MAIT. J Immunol. 2014;192(10):4475–80. PubMed Central PMCID: PMCPMC4025967. pmid:24795465
  50. 50. Napier RJ, Adams EJ, Gold MC, Lewinsohn DM. The Role of Mucosal Associated Invariant T Cells in Antimicrobial Immunity. Front Immunol. 2015;6:344. PubMed Central PMCID: PMCPMC4492155. pmid:26217338
  51. 51. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401(6754):708–12. pmid:10537110
  52. 52. Larbi A, Mitjavila-Garcia MT, Flamant S, Valogne Y, Clay D, Usunier B, et al. Generation of multipotent early lymphoid progenitors from human embryonic stem cells. Stem cells and development. 2014;23(24):2983–95. pmid:24955741
  53. 53. Neuhaus J, Jacobs DR Jr., Baker JV, Calmy A, Duprez D, La Rosa A, et al. Markers of inflammation, coagulation, and renal function are elevated in adults with HIV infection. The Journal of infectious diseases. 2010;201(12):1788–95. Epub 2010/05/08. PubMed Central PMCID: PMC2872049. pmid:20446848
  54. 54. Hearps AC, Martin GE, Rajasuriar R, Crowe SM. Inflammatory co-morbidities in HIV+ individuals: learning lessons from healthy ageing. Curr HIV/AIDS Rep. 2014;11(1):20–34. pmid:24414166
  55. 55. Klatt NR, Funderburg NT, Brenchley JM. Microbial translocation, immune activation, and HIV disease. Trends Microbiol. 2013;21(1):6–13. PubMed Central PMCID: PMCPMC3534808. pmid:23062765
  56. 56. Le Bourhis L, Martin E, Peguillet I, Guihot A, Froux N, Core M, et al. Antimicrobial activity of mucosal-associated invariant T cells. Nat Immunol. 2010;11(8):701–8. pmid:20581831
  57. 57. Leeansyah E, Ganesh A, Quigley MF, Sonnerborg A, Andersson J, Hunt PW, et al. Activation, exhaustion, and persistent decline of the antimicrobial MR1-restricted MAIT-cell population in chronic HIV-1 infection. Blood. 2013;121(7):1124–35. PubMed Central PMCID: PMCPMC3575756. pmid:23243281
  58. 58. Serrano-Villar S, Sainz T, Lee SA, Hunt PW, Sinclair E, Shacklett BL, et al. HIV-infected individuals with low CD4/CD8 ratio despite effective antiretroviral therapy exhibit altered T cell subsets, heightened CD8+ T cell activation, and increased risk of non-AIDS morbidity and mortality. PLoS Pathog. 2014;10(5):e1004078. PubMed Central PMCID: PMCPMC4022662. pmid:24831517
  59. 59. Velu V, Titanji K, Zhu B, Husain S, Pladevega A, Lai L, et al. Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature. 2009;458(7235):206–10. PubMed Central PMCID: PMCPMC2753387. pmid:19078956
  60. 60. Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10(1):29–37. PubMed Central PMCID: PMCPMC2605166. pmid:19043418
  61. 61. Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med. 2009;15(8):893–900. PubMed Central PMCID: PMCPMC2859814. pmid:19543283
  62. 62. Brown EJ, Albers MW, Shin TB, Ichikawa K, Keith CT, Lane WS, et al. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature. 1994;369(6483):756–8. pmid:8008069
  63. 63. Chiu MI, Katz H, Berlin V. RAPT1, a mammalian homolog of yeast Tor, interacts with the FKBP12/rapamycin complex. Proc Natl Acad Sci U S A. 1994;91(26):12574–8. PubMed Central PMCID: PMCPMC45481. pmid:7809080